55 results on '"Graham McClorey"'
Search Results
2. Uniform sarcolemmal dystrophin expression is required to prevent extracellular microRNA release and improve dystrophic pathology
- Author
-
Tirsa L.E. vanWestering, Yulia Lomonosova, Anna M.L. Coenen‐Stass, Corinne A. Betts, Amarjit Bhomra, Margriet Hulsker, Lucy E. Clark, Graham McClorey, Annemieke Aartsma‐Rus, Maaike vanPutten, Matthew J.A. Wood, and Thomas C. Roberts
- Subjects
Duchenne muscular dystrophy ,MicroRNA ,Biomarkers ,Dystrophin ,X‐chromosome inactivation ,Muscle turnover ,Diseases of the musculoskeletal system ,RC925-935 ,Human anatomy ,QM1-695 - Abstract
Abstract Background Duchenne muscular dystrophy (DMD) is a fatal muscle‐wasting disorder caused by genetic loss of dystrophin protein. Extracellular microRNAs (ex‐miRNAs) are putative, minimally invasive biomarkers of DMD. Specific ex‐miRNAs (e.g. miR‐1, miR‐133a, miR‐206, and miR‐483) are highly up‐regulated in the serum of DMD patients and dystrophic animal models and are restored to wild‐type levels following exon skipping‐mediated dystrophin rescue in mdx mice. As such, ex‐miRNAs are promising pharmacodynamic biomarkers of exon skipping efficacy. Here, we aimed to determine the degree to which ex‐miRNA levels reflect the underlying level of dystrophin protein expression in dystrophic muscle. Methods Candidate ex‐miRNA biomarker levels were investigated in mdx mice in which dystrophin was restored with peptide‐PMO (PPMO) exon skipping conjugates and in mdx‐XistΔhs mice that express variable amounts of dystrophin from birth as a consequence of skewed X‐chromosome inactivation. miRNA profiling was performed in mdx‐XistΔhs mice using the FirePlex methodology and key results validated by small RNA TaqMan RT‐qPCR. The muscles from each animal model were further characterized by dystrophin western blot and immunofluorescence staining. Results The restoration of ex‐myomiR abundance observed following PPMO treatment was not recapitulated in the high dystrophin‐expressing mdx‐XistΔhs group, despite these animals expressing similar amounts of total dystrophin protein (~37% of wild‐type levels). Instead, ex‐miRNAs were present at high levels in mdx‐XistΔhs mice regardless of dystrophin expression. PPMO‐treated muscles exhibited a uniform pattern of dystrophin localization and were devoid of regenerating fibres, whereas mdx‐XistΔhs muscles showed non‐homogeneous dystrophin staining and sporadic regenerating foci. Conclusions Uniform dystrophin expression is required to prevent ex‐miRNA release, stabilize myofiber turnover, and attenuate pathology in dystrophic muscle.
- Published
- 2020
- Full Text
- View/download PDF
3. Peptide-conjugated phosphodiamidate oligomer-mediated exon skipping has benefits for cardiac function in mdx and Cmah-/-mdx mouse models of Duchenne muscular dystrophy.
- Author
-
Alison M Blain, Elizabeth Greally, Graham McClorey, Raquel Manzano, Corinne A Betts, Caroline Godfrey, Liz O'Donovan, Thibault Coursindel, Mike J Gait, Matthew J Wood, Guy A MacGowan, and Volker W Straub
- Subjects
Medicine ,Science - Abstract
Cardiac failure is a major cause of mortality in patients with Duchenne muscular dystrophy (DMD). Antisense-mediated exon skipping has the ability to correct out-of-frame mutations in DMD to produce truncated but functional dystrophin. Traditional antisense approaches have however been limited by their poor uptake into cardiac muscle. The addition of cell-penetrating peptides to antisense molecules has increased their potency and improved their uptake into all muscles, including the heart. We have investigated the efficacy of the Peptide-conjugated phosphodiamidate morpholino oligomer (P-PMO) Pip6a-PMO, for restoration of cardiac dystrophin and functional rescue in DMD mice- the mdx mouse and the less well characterised Cmah-/-mdx mouse (which carry a human-like mutation in the mouse Cmah gene as well as a mutation in DMD). In our first study male mdx mice were administered Pip6a-PMO, i.v, fortnightly from 12 to 30 weeks of age alongside mock-injected age-matched mdx and C57BL10 controls. Mice received 4 doses of 18 mg/kg followed by 8 doses of 12.5 mg/kg. The cardiac function of the mice was analysed 2 weeks after their final injection by MRI followed by conductance catheter and their muscles were harvested for dystrophin quantification. In the second study, male Cmah-/-mdx mice, received 12.5 mg/kg Pip6a-PMO, i.v fortnightly from 8 to 26 weeks and assessed by MRI at 3 time points (12, 18 and 28 weeks) alongside mock-injected age-matched mdx, C57BL10 and Cmah-/-mdx controls. The mice also underwent MEMRI and conductance catheter at 28 weeks. This allowed us to characterise the cardiac phenotype of Cmah-/-mdx mice as well as assess the effects of P-PMO on cardiac function. Pip6a-PMO treatment resulted in significant restoration of dystrophin in mdx and Cmah-/-mdx mice (37.5% and 51.6%, respectively), which was sufficient to significantly improve cardiac function, ameliorating both right and left ventricular dysfunction. Cmah-/-mdx mice showed an abnormal response to dobutamine stress test and this was completely ameliorated by PIP6a-PMO treatment. These encouraging data suggest that total restoration of dystrophin may not be required to significantly improve cardiac outcome in DMD patients and that it may be realistic to expect functional improvements with modest levels of dystrophin restoration which may be very achievable in future clinical trials.
- Published
- 2018
- Full Text
- View/download PDF
4. Correlating In Vitro Splice Switching Activity With Systemic In Vivo Delivery Using Novel ZEN-modified Oligonucleotides
- Author
-
Suzan M Hammond, Graham McClorey, Joel Z Nordin, Caroline Godfrey, Sofia Stenler, Kim A Lennox, CI Edvard Smith, Ashley M Jacobi, Miguel A Varela, Yi Lee, Mark A Behlke, Matthew J A Wood, and Samir E L Andaloussi
- Subjects
antisense oligonucleotides ,Duchenne muscular dystrophy ,splice switching ,ZEN modification ,Therapeutics. Pharmacology ,RM1-950 - 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.
- Published
- 2014
- Full Text
- View/download PDF
5. Expression Analysis in Multiple Muscle Groups and Serum Reveals Complexity in the MicroRNA Transcriptome of the mdx Mouse with Implications for Therapy
- Author
-
Thomas C Roberts, K Emelie M Blomberg, Graham McClorey, Samir EL Andaloussi, Caroline Godfrey, Corinne Betts, Thibault Coursindel, Michael J Gait, CI Edvard Smith, and Matthew JA Wood
- Subjects
Duchenne muscular dystrophy ,mdx ,microarray ,microRNA ,Therapeutics. Pharmacology ,RM1-950 - Abstract
MicroRNAs (miRNAs) are a class of small RNAs that regulate gene expression and are implicated in wide-ranging cellular processes and pathological conditions including Duchenne muscular dystrophy (DMD). We have compared differential miRNA expression in proximal and distal limb muscles, diaphragm, heart and serum in the mdx mouse relative to wild-type controls. Global transcriptome analysis revealed muscle-specific patterns of differential miRNA expression as well as a number of changes common between tissues, including previously identified dystromirs. In the case of miR-31 and miR-34c, upregulation of primary-miRNA transcripts, precursor hairpins and all mature miRNAs derived from the same transcript or miRNA cluster, strongly suggests transcriptional regulation of these miRNAs. The most striking differences in differential miRNA expression were between muscle tissue and serum. Specifically, miR-1, miR-133a, and miR-206 were highly abundant in mdx serum but downregulated or modestly upregulated in muscle, suggesting that these miRNAs are promising disease biomarkers. Indeed, the relative serum levels of these miRNAs were normalized in response to peptide-phosphorodiamidate morpholino oligonucleotide (PMO) mediated dystrophin restoration therapy. This study has revealed further complexity in the miRNA transcriptome of the mdx mouse, an understanding of which will be valuable in the development of novel therapeutics and for monitoring their efficacy.
- Published
- 2012
- Full Text
- View/download PDF
6. Dual Myostatin and Dystrophin Exon Skipping by Morpholino Nucleic Acid Oligomers Conjugated to a Cell-penetrating Peptide Is a Promising Therapeutic Strategy for the Treatment of Duchenne Muscular Dystrophy
- Author
-
Alberto Malerba, Jagjeet K Kang, Graham McClorey, Amer F Saleh, Linda Popplewell, Michael J Gait, Matthew JA Wood, and George Dickson
- Subjects
antisense oligonucleotides ,Duchenne muscular dystrophy ,dystrophin ,exon skipping ,myostatin ,Therapeutics. Pharmacology ,RM1-950 - Abstract
The knockdown of myostatin, a negative regulator of skeletal muscle mass may have important implications in disease conditions accompanied by muscle mass loss like cancer, HIV/AIDS, sarcopenia, muscle atrophy, and Duchenne muscular dystrophy (DMD). In DMD patients, where major muscle loss has occurred due to a lack of dystrophin, the therapeutic restoration of dystrophin expression alone in older patients may not be sufficient to restore the functionality of the muscles. We recently demonstrated that phosphorodiamidate morpholino oligomers (PMOs) can be used to re-direct myostatin splicing and promote the expression of an out-of-frame transcript so reducing the amount of the synthesized myostatin protein. Furthermore, the systemic administration of the same PMO conjugated to an octaguanidine moiety (Vivo-PMO) led to a significant increase in the mass of soleus muscle of treated mice. Here, we have further optimized the use of Vivo-PMO in normal mice and also tested the efficacy of the same PMO conjugated to an arginine-rich cell-penetrating peptide (B-PMO). Similar experiments conducted in mdx dystrophic mice showed that B-PMO targeting myostatin is able to significantly increase the tibialis anterior (TA) muscle weight and when coadministered with a B-PMO targeting the dystrophin exon 23, it does not have a detrimental interaction. This study confirms that myostatin knockdown by exon skipping is a potential therapeutic strategy to counteract muscle wasting conditions and dual myostatin and dystrophin skipping has potential as a therapy for DMD.
- Published
- 2012
- Full Text
- View/download PDF
7. Small RNA-Mediated Epigenetic Myostatin Silencing
- Author
-
Thomas C Roberts, Samir EL Andaloussi, Kevin V Morris, Graham McClorey, and Matthew JA Wood
- Subjects
epigenetic ,muscle degeneration ,myostatin ,siRNA ,transcriptional gene silencing ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Myostatin (Mstn) is a secreted growth factor that negatively regulates muscle mass and is therefore a potential pharmacological target for the treatment of muscle wasting disorders such as Duchenne muscular dystrophy. Here we describe a novel Mstn blockade approach in which small interfering RNAs (siRNAs) complementary to a promoter-associated transcript induce transcriptional gene silencing (TGS) in two differentiated mouse muscle cell lines. Silencing is sensitive to treatment with the histone deacetylase inhibitor trichostatin A, and the silent state chromatin mark H3K9me2 is enriched at the Mstn promoter following siRNA transfection, suggesting epigenetic remodeling underlies the silencing effect. These observations suggest that long-term epigenetic silencing may be feasible for Mstn and that TGS is a promising novel therapeutic strategy for the treatment of muscle wasting disorders.
- Published
- 2012
- Full Text
- View/download PDF
8. Modulating the expression of disease genes with RNA-based therapy.
- Author
-
Matthew Wood, Haifang Yin, and Graham McClorey
- Subjects
Genetics ,QH426-470 - Abstract
Conventional gene therapy has focused largely on gene replacement in target cells. However, progress from basic research to the clinic has been slow for reasons relating principally to the challenges of heterologous DNA delivery and regulation in vivo. Alternative approaches targeting RNA have the potential to circumvent some of these difficulties, particularly as the active therapeutic molecules are usually short oligonucleotides and the target gene transcript is under endogenous regulation. RNA-based strategies offer a series of novel therapeutic applications, including altered processing of the target pre-mRNA transcript, reprogramming of genetic defects through mRNA repair, and the targeted silencing of allele- or isoform-specific gene transcripts. This review examines the potential of RNA therapeutics, focusing on antisense oligonucleotide modification of pre-mRNA splicing, methods for pre-mRNA trans-splicing, and the isoform- and allele-specific applications of RNA interference.
- Published
- 2007
- Full Text
- View/download PDF
9. Exon skipping induces uniform dystrophin rescue with dose-dependent restoration of serum miRNA biomarkers and muscle biophysical properties
- Author
-
Katarzyna Chwalenia, Jacopo Oieni, Joanna Zemła, Małgorzata Lekka, Nina Ahlskog, Anna M.L. Coenen-Stass, Graham McClorey, Matthew J.A. Wood, Yulia Lomonosova, and Thomas C. Roberts
- Subjects
Drug Discovery ,Molecular Medicine - Abstract
Therapies that restore dystrophin expression are presumed to correct Duchenne muscular dystrophy (DMD), with antisense-mediated exon skipping being the leading approach. Here we aimed to determine whether exon skipping using a peptide-phosphorodiamidate morpholino oligonucleotide (PPMO) conjugate results in dose-dependent restoration of uniform dystrophin localization, together with correction of putative DMD serum and muscle biomarkers. Dystrophin-deficient
- Published
- 2022
10. Oligonucleotide analogues with locked-amide linkages have therapeutic potential
- Author
-
Ysobel Baker, Cameron Thorpe, Jinfeng Chen, Laura Poller, Lina Cox, Pawan Kumar, Wooi Fang Lim, Lillian Lie, Graham McClorey, Sven Epple, Daniel Singleton, Michael McDonough, Jack Hardwick, Kirsten Christensen, Matthew Wood, James Hall, Afaf El-Sagheer, and Tom Brown
- Abstract
Oligonucleotides that target mRNA have great promise as therapeutic agents for life-threatening conditions but suffer from poor bioavailability, hence high cost. As currently untreatable diseases come within the reach of oligonucleotide therapies, new analogues are urgently needed to address this. With this in mind we have developed reduced-charge oligonucleotides containing artificial LNA-amide linkages with improved gymnotic cell uptake, RNA affinity, stability and potency. To construct such oligonucleotides, five LNA-amide monomers (A, T, C, 5mC and G), where the 3´-OH is replaced by an ethanoic acid group, were synthesised in good yield and used in solid-phase oligonucleotide synthesis to form amide linkages with high efficiency. The artificial backbone causes minimal structural deviation to the DNA:RNA duplex. These studies indicate that splice-switching oligonucleotides containing LNA-amide linkages and phosphorothioates display improved activity relative to oligonucleotides lacking amides, highlighting the therapeutic potential of this technology.
- Published
- 2022
11. Control of backbone chemistry and chirality boost oligonucleotide splice switching activity
- Author
-
Pachamuthu Kandasamy, Graham McClorey, Mamoru Shimizu, Nayantara Kothari, Rowshon Alam, Naoki Iwamoto, Jayakanthan Kumarasamy, Gopal R Bommineni, Adam Bezigian, Onanong Chivatakarn, David C D Butler, Michael Byrne, Katarzyna Chwalenia, Kay E Davies, Jigar Desai, Juili Dilip Shelke, Ann F Durbin, Ruth Ellerington, Ben Edwards, Jack Godfrey, Andrew Hoss, Fangjun Liu, Kenneth Longo, Genliang Lu, Subramanian Marappan, Jacopo Oieni, Ik-Hyeon Paik, Erin Purcell Estabrook, Chikdu Shivalila, Maeve Tischbein, Tomomi Kawamoto, Carlo Rinaldi, Joana Rajão-Saraiva, Snehlata Tripathi, Hailin Yang, Yuan Yin, Xiansi Zhao, Cong Zhou, Jason Zhang, Luciano Apponi, Matthew J A Wood, and Chandra Vargeese
- Subjects
Muscular Dystrophy, Duchenne ,Mice ,RNA Splicing ,Genetics ,Animals ,Phosphorothioate Oligonucleotides ,Exons ,Oligonucleotides, Antisense ,Muscle, Skeletal - Abstract
Although recent regulatory approval of splice-switching oligonucleotides (SSOs) for the treatment of neuromuscular disease such as Duchenne muscular dystrophy has been an advance for the splice-switching field, current SSO chemistries have shown limited clinical benefit due to poor pharmacology. To overcome limitations of existing technologies, we engineered chimeric stereopure oligonucleotides with phosphorothioate (PS) and phosphoryl guanidine-containing (PN) backbones. We demonstrate that these chimeric stereopure oligonucleotides have markedly improved pharmacology and efficacy compared with PS-modified oligonucleotides, preventing premature death and improving median survival from 49 days to at least 280 days in a dystrophic mouse model with an aggressive phenotype. These data demonstrate that chemical optimization alone can profoundly impact oligonucleotide pharmacology and highlight the potential for continued innovation around the oligonucleotide backbone. More specifically, we conclude that chimeric stereopure oligonucleotides are a promising splice-switching modality with potential for the treatment of neuromuscular and other genetic diseases impacting difficult to reach tissues such as the skeletal muscle and heart.
- Published
- 2022
12. Dystrophin involvement in peripheral circadian SRF signalling
- Author
-
Wooi F. Lim, Melissa Bowerman, Corinne A. Betts, Lara Cravo, Aarti Jagannath, Tirsa L.E. van Westering, Jinhong Meng, Katarzyna Chwalenia, Russell G. Foster, Jennifer E. Morgan, Carlo Rinaldi, Maria Sofia Falzarano, Elizabeth O’Donovan, Alessandra Ferlini, Graham McClorey, Katharina E. Meijboom, John R. Counsell, Amarjit Bhomra, Matthew J.A. Wood, Michael J. Gait, Amer F. Saleh, and Subhashis Banerjee
- Subjects
Serum Response Factor ,genetic structures ,Utrophin ,Myoblasts, Skeletal ,Health, Toxicology and Mutagenesis ,Plant Science ,Biochemistry, Genetics and Molecular Biology (miscellaneous) ,Cell Line ,Dystrophin ,Mice ,Mediator ,Transcription (biology) ,medicine ,Animals ,Circadian rhythm ,Muscular dystrophy ,Research Articles ,Ecology ,biology ,Myogenesis ,Suprachiasmatic nucleus ,musculoskeletal system ,medicine.disease ,eye diseases ,Actins ,Cell biology ,embryonic structures ,cardiovascular system ,biology.protein ,medicine.symptom ,rhoA GTP-Binding Protein ,RC ,Signal Transduction ,Research Article ,Muscle contraction - Abstract
Absence of integral sarcolemmal protein, dystrophin, disrupts the RhoA-actin-SRF cascade in skeletal muscle, with subsequent dysregulation of downstream-SRF circadian targets and circadian rhythm., 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.
- Published
- 2021
13. Dystrophin regulates peripheral circadian SRF signalling
- Author
-
Katarzyna Chwalenia, O'Donovan E, Russell G. Foster, Carlo Rinaldi, Graham McClorey, Soumya Banerjee, John R. Counsell, Jennifer E. Morgan, van Westering T, Saleh A, Lara Cravo, Jinhong Meng, Maria Sofia Falzarano, Melissa Bowerman, Matthew J.A. Wood, Corinne A. Betts, Lim C, Aarti Jagannath, Amarjit Bhomra, Gait Mj, Katharina E. Meijboom, and Ferlini A
- Subjects
musculoskeletal diseases ,Myogenesis ,Suprachiasmatic nucleus ,Biology ,musculoskeletal system ,medicine.disease ,Cell biology ,Mediator ,Transcription (biology) ,biology.protein ,medicine ,Circadian rhythm ,medicine.symptom ,Muscular dystrophy ,Dystrophin ,Muscle contraction - Abstract
Dystrophin is a sarcolemmal protein essential for muscle contraction and maintenance, absence of which leads to the devastating muscle wasting disease Duchenne muscular dystrophy (DMD)[1, 2]. Dystrophin has an actin-binding domain [3–5], which specifically binds and stabilises filamentous (F)-actin[6], an integral component of the RhoA-actin-serum response factor (SRF)-pathway[7]. The RhoA-actin-SRF-pathway plays an essential role in circadian signalling whereby the hypothalamic suprachiasmatic nucleus, transmits systemic cues to peripheral tissues, activating SRF and transcription of clock target genes[8, 9]. Given dystrophin binds F-actin and disturbed SRF-signalling disrupts clock entrainment, we hypothesised that dystrophin loss causes circadian deficits. Here we show for the first time alterations in the RhoA-actin-SRF-signalling-pathway, in both dystrophin-deficient myotubes and dystrophic mouse models. Specifically, we demonstrate reduced F/G-actin ratios and nuclear MRTF, dysregulation of core clock and downstream target-genes, and down-regulation of key circadian genes in muscle biopsies from DMD patients harbouring an array of mutations. Further, disrupted circadian locomotor behaviour was observed in dystrophic mice indicative of disrupted SCN signalling, and indeed dystrophin protein was absent in the SCN of dystrophic animals. Dystrophin is thus a critically important component of the RhoA-actin-SRF-pathway and a novel mediator of circadian signalling in peripheral tissues, loss of which leads to circadian dysregulation.
- Published
- 2021
14. Fine Tuning of Phosphorothioate Inclusion in 2′-O-Methyl Oligonucleotides Contributes to Specific Cell Targeting for Splice-Switching Modulation
- Author
-
Aoki, Yoshitsugu, primary, Rocha, Cristina S. J., additional, Lehto, Taavi, additional, Miyatake, Shouta, additional, Johansson, Henrik, additional, Hashimoto, Yasumasa, additional, Nordin, Joel Z., additional, Mager, Imre, additional, Aoki, Misako, additional, Graham, McClorey, additional, Sathyaprakash, Chaitra, additional, Roberts, Thomas C., additional, Wood, Matthew J. A., additional, Behlke, Mark A., additional, and Andaloussi, Samir El, additional
- Published
- 2021
- Full Text
- View/download PDF
15. Immortalized Canine Dystrophic Myoblast Cell Lines for Development of Peptide-Conjugated Splice-Switching Oligonucleotides
- Author
-
Yoshitsugu Aoki, A Arzumanov, Kamel Mamchaoui, Tone Yuichiro, Rika Maruyama, Graham McClorey, Shin'ichi Takeda, Toshifumi Yokota, Yasumasa Hashimoto, Maria K. Tsoumpra, Michihiro Imamura, Vincent Mouly, Michael J. Gait, Matthew J.A. Wood, Reiko Terada, Gestionnaire, HAL Sorbonne Université 5, Department of Neuromuscular Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Centre de recherche en Myologie – U974 SU-INSERM, Institut National de la Santé et de la Recherche Médicale (INSERM)-Sorbonne Université (SU), University of Alberta, University of Oxford, UMRS974, Université Pierre et Marie Curie - Paris 6 (UPMC), University of Oxford [Oxford], Institut de Myologie, Centre National de la Recherche Scientifique (CNRS)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Association française contre les myopathies (AFM-Téléthon)-Sorbonne Université (SU), and Centre de Recherche en Myologie
- Subjects
0301 basic medicine ,canine X-linked muscular dystrophy in Japan (CXMD J ) ,Duchenne muscular dystrophy ,Biochemistry ,Morpholinos ,Dystrophin ,Myoblasts ,Mice ,0302 clinical medicine ,Drug Discovery ,Muscular dystrophy ,Telomerase ,ComputingMilieux_MISCELLANEOUS ,biology ,Chemistry ,Myogenesis ,phosphorodiamidate morpholino oligomer ,Exons ,Original Papers ,3. Good health ,Cell biology ,030220 oncology & carcinogenesis ,Molecular Medicine ,[SDV.MHEP.AHA] Life Sciences [q-bio]/Human health and pathology/Tissues and Organs [q-bio.TO] ,splice-switching oligonucleotides ,Nonsense mutation ,[SDV.BC]Life Sciences [q-bio]/Cellular Biology ,Cell Line ,03 medical and health sciences ,Dogs ,Genetics ,medicine ,[SDV.MHEP.AHA]Life Sciences [q-bio]/Human health and pathology/Tissues and Organs [q-bio.TO] ,Animals ,Humans ,Telomerase reverse transcriptase ,Molecular Biology ,Cyclin-Dependent Kinase 4 ,Genetic Therapy ,immortalized dystrophic canine myoblast ,Oligonucleotides, Antisense ,canine X-linked muscular dystrophy in Japan (CXMDJ) ,medicine.disease ,Exon skipping ,Muscular Dystrophy, Duchenne ,030104 developmental biology ,biology.protein ,Cell-penetrating peptide ,RNA Splice Sites ,Peptides ,cell-penetrating peptide - 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.
- Published
- 2021
16. Engineered extracellular vesicle decoy receptor-mediated modulation of the IL6 trans-signalling pathway in muscle
- Author
-
Oscar P. B. Wiklander, Dhanu Gupta, Thomas C. Roberts, André Görgens, Matthew J.A. Wood, Samir El Andaloussi, Joel Z. Nordin, Per Lundin, Mariana Conceição, Antonio Musarò, Laura Forcina, Imre Mäger, and Graham McClorey
- Subjects
Chemistry ,Myogenesis ,Receptor-mediated endocytosis ,Extracellular vesicle ,Decoy receptors ,skin and connective tissue diseases ,Receptor ,Decoy ,C2C12 ,biological factors ,Hedgehog signaling pathway ,Cell biology - Abstract
The cytokine interleukin 6 (IL6) is a key mediator of inflammation that contributes to skeletal muscle pathophysiology. IL6 activates target cells by two different mechanisms, the classical and transsignalling pathways. While classical signalling is associated with the anti-inflammatory activities of the cytokine, the IL6 trans-signalling pathway mediates chronic inflammation and is therefore a target for therapeutic intervention. Extracellular vesicles (EVs) are natural, lipid-bound nanoparticles, with potential as targeted delivery vehicles for therapeutic macromolecules. Here, we engineered EVs to express IL6 signal transducer (IL6ST) decoy receptors to selectively inhibit the IL6 trans-signalling pathway. The potency of the IL6ST decoy receptor EVs was optimized by inclusion of a GCN4 dimerization domain and a peptide sequence derived from syntenin-1 which targets the decoy receptor to EVs. The resulting engineered EVs were able to efficiently inhibit activation of the IL6 transsignalling pathway in reporter cells, while having no effect on the IL6 classical signalling. IL6ST decoy receptor EVs, were also capable of blocking the IL6 trans-signalling pathway in C2C12 myoblasts and myotubes, thereby inhibiting the phosphorylation of STAT3 and partially reversing the anti-differentiation effects observed when treating cells with IL6/IL6R complexes. Treatment of a Duchenne muscular dystrophy mouse model with IL6ST decoy receptor EVs resulted in a reduction in STAT3 phosphorylation in the quadriceps and gastrocnemius muscles of these mice, thereby demonstrating in vivo activity of the decoy receptor EVs as a potential therapy. Taken together, this study reveals the IL6 trans-signalling pathway as a promising therapeutic target in DMD, and demonstrates the therapeutic potential of IL6ST decoy receptor EVs.
- Published
- 2020
17. Uniform sarcolemmal dystrophin expression is required to prevent extracellular microRNA release and improve dystrophic pathology
- Author
-
Lucy E Clark, Yulia Lomonosova, Amarjit Bhomra, Annemieke Aartsma-Rus, Anna M.L. Coenen-Stass, Margriet Hulsker, Tirsa L.E. van Westering, Thomas C. Roberts, Graham McClorey, Corinne A. Betts, Matthew J.A. Wood, and Maaike van Putten
- Subjects
0301 basic medicine ,musculoskeletal diseases ,Duchenne muscular dystrophy ,Pathology ,medicine.medical_specialty ,congenital, hereditary, and neonatal diseases and abnormalities ,lcsh:Diseases of the musculoskeletal system ,lcsh:QM1-695 ,Dystrophin ,Mice ,03 medical and health sciences ,Exon ,Sarcolemma ,0302 clinical medicine ,Western blot ,X‐chromosome inactivation ,Physiology (medical) ,microRNA ,medicine ,Extracellular ,Animals ,Humans ,Orthopedics and Sports Medicine ,Child ,medicine.diagnostic_test ,biology ,business.industry ,X-chromosome inactivation ,MicroRNA ,Original Articles ,lcsh:Human anatomy ,medicine.disease ,musculoskeletal system ,Exon skipping ,Disease Models, Animal ,MicroRNAs ,030104 developmental biology ,030220 oncology & carcinogenesis ,biology.protein ,Biomarker (medicine) ,Original Article ,Female ,lcsh:RC925-935 ,business ,Muscle turnover ,Biomarkers - Abstract
Background:Duchenne muscular dystrophy (DMD) is a fatal muscle‐wasting disorder caused by genetic loss of dystrophin protein. Extracellular microRNAs (ex‐miRNAs) are putative, minimally invasive biomarkers of DMD. Specific ex‐miRNAs (e.g. miR‐1, miR‐133a, miR‐206, and miR‐483) are highly up‐regulated in the serum of DMD patients and dystrophic animal models and are restored to wild‐type levels following exon skipping‐mediated dystrophin rescue inmdxmice. As such, ex‐miRNAs are promising pharmacodynamic biomarkers of exon skipping efficacy. Here, we aimed to determine the degree to which ex‐miRNA levels reflect the underlying level of dystrophin protein expression in dystrophic muscle. Methods:Candidate ex‐miRNA biomarker levels were investigated inmdxmice in which dystrophin was restored with peptide‐PMO (PPMO) exon skipping conjugates and inmdx‐XistΔhsmice that express variable amounts of dystrophin from birth as a consequence of skewed X‐chromosome inactivation. miRNA profiling was performed inmdx‐XistΔhsmice using the FirePlex methodology and key results validated by small RNA TaqMan RT‐qPCR. The muscles from each animal model were further characterized by dystrophin western blot and immunofluorescence staining. Results:The restoration of ex‐myomiR abundance observed following PPMO treatment was not recapitulated in the high dystrophin‐expressingmdx‐XistΔhsgroup, despite these animals expressing similar amounts of total dystrophin protein (~37% of wild‐type levels). Instead, ex‐miRNAs were present at high levels inmdx‐XistΔhsmice regardless of dystrophin expression. PPMO‐treated muscles exhibited a uniform pattern of dystrophin localization and were devoid of regenerating fibres, whereasmdx‐XistΔhsmuscles showed non‐homogeneous dystrophin staining and sporadic regenerating foci. Conclusions:Uniform dystrophin expression is required to prevent ex‐miRNA release, stabilize myofiber turnover, and attenuate pathology in dystrophic muscle.
- Published
- 2019
18. Profile of circadianly regulated metabolic genes in dystrophic heart
- Author
-
Melissa Bowerman, K Meijboom, Corinne A. Betts, Wood Mja., van Westering Tle., and Graham McClorey
- Subjects
medicine.medical_specialty ,biology ,business.industry ,Duchenne muscular dystrophy ,Cardiomyopathy ,Disease ,Carbohydrate metabolism ,medicine.disease ,Endocrinology ,Neurology ,Internal medicine ,Diabetes mellitus ,Pediatrics, Perinatology and Child Health ,biology.protein ,medicine ,Neurology (clinical) ,Circadian rhythm ,medicine.symptom ,Dystrophin ,business ,Wasting ,Genetics (clinical) - Abstract
Duchenne muscular dystrophy (DMD) is a monogenic disorder caused by the lack of the integral structural protein, dystrophin, which results in severe muscle wasting and cardiomyopathy in affected boys. Indeed, cardiorespiratory complications are the predominant cause for mortality in DMD patients. We have recently shown that circadian rhythm is disrupted in dystrophic mice as a direct result of the lack of dystrophin protein. It is well reported that disruption of circadian rhythmicity leads to perturbed metabolism and an array of disorders including obesity, diabetes and cardiovascular disease. Disturbed cardiac metabolism in DMD patients and dystrophic mice is also well described, and thus it would be interesting to learn whether pertinent metabolic genes which are known to be circadianly regulated, are disrupted in dystrophic mice. Here we show for the first time, significant changes in the differential expression patterns of multiple genes involved in free fatty acid and glucose metabolism, in 2 mouse models of DMD compared to control mice. These findings provide the foundation for further research to better understand the metabolic/circadian milieu and its effect on dystrophic heart, so that we may devise strategies to augment cardiac metabolism, in an effort to halt the deterioration in cardiac phenotype.
- Published
- 2018
19. Cell-penetrating peptides to enhance delivery of Oligonucleotide-based therapeutics
- Author
-
Graham McClorey and Subhashis Banerjee
- Subjects
cell-penetrating peptides ,0301 basic medicine ,Oligonucleotide ,Chemistry ,Cell ,Medicine (miscellaneous) ,Translation (biology) ,Review ,General Biochemistry, Genetics and Molecular Biology ,Bioavailability ,03 medical and health sciences ,030104 developmental biology ,medicine.anatomical_structure ,Biochemistry ,Antisense oligonucleotides ,Nucleic acid ,Systemic administration ,medicine ,antisense oligonucleotides ,delivery - Abstract
The promise of nucleic acid based oligonucleotides as effective genetic therapies has been held back by their low bioavailability and poor cellular uptake to target tissues upon systemic administration. One such strategy to improve upon delivery is the use of short cell-penetrating peptides (CPPs) that can be either directly attached to their cargo through covalent linkages or through the formation of noncovalent nanoparticle complexes that can facilitate cellular uptake. In this review, we will highlight recent proof-of-principle studies that have utilized both of these strategies to improve nucleic acid delivery and discuss the prospects for translation of this approach for clinical application.
- Published
- 2018
20. Peptide-conjugated phosphodiamidate oligomer-mediated exon skipping has benefits for cardiac function in mdx and Cmah-/-mdx mouse models of Duchenne muscular dystrophy
- Author
-
Raquel Manzano, A. Blain, M J Gait, Matthew J.A. Wood, Caroline Godfrey, Liz O'Donovan, Volker Straub, Corinne A. Betts, Guy A. MacGowan, Thibault Coursindel, E. Greally, and Graham McClorey
- Subjects
0301 basic medicine ,mdx mouse ,Heredity ,Morpholino ,Genetic Linkage ,Duchenne muscular dystrophy ,lcsh:Medicine ,Duchenne Muscular Dystrophy ,Cell-Penetrating Peptides ,030204 cardiovascular system & hematology ,Biochemistry ,Muscular Dystrophies ,Morpholinos ,Dystrophin ,Mice ,0302 clinical medicine ,Medicine and Health Sciences ,Muscular dystrophy ,lcsh:Science ,Frameshift Mutation ,Musculoskeletal System ,Multidisciplinary ,biology ,Muscles ,Cardiac muscle ,Heart ,Animal Models ,Exons ,musculoskeletal system ,Chemistry ,medicine.anatomical_structure ,Experimental Organism Systems ,Neurology ,X-Linked Traits ,Sex Linkage ,Physical Sciences ,Anatomy ,Research Article ,Chemical Elements ,Biotechnology ,Cardiac function curve ,musculoskeletal diseases ,medicine.medical_specialty ,congenital, hereditary, and neonatal diseases and abnormalities ,Catheters ,Mouse Models ,Research and Analysis Methods ,03 medical and health sciences ,Model Organisms ,Internal medicine ,Genetics ,medicine ,Animals ,Cardiac Muscles ,Clinical Genetics ,Heart Failure ,Manganese ,business.industry ,Myocardium ,lcsh:R ,Biology and Life Sciences ,Proteins ,medicine.disease ,Fibrosis ,Exon skipping ,Muscular Dystrophy, Duchenne ,Cytoskeletal Proteins ,Disease Models, Animal ,030104 developmental biology ,Endocrinology ,Cardiovascular Anatomy ,Mice, Inbred mdx ,biology.protein ,lcsh:Q ,Medical Devices and Equipment ,business ,Developmental Biology - Abstract
Cardiac failure is a major cause of mortality in patients with Duchenne muscular dystrophy (DMD). Antisense-mediated exon skipping has the ability to correct out-of-frame mutations in DMD to produce truncated but functional dystrophin. Traditional antisense approaches have however been limited by their poor uptake into cardiac muscle. The addition of cell-penetrating peptides to antisense molecules has increased their potency and improved their uptake into all muscles, including the heart. We have investigated the efficacy of the Peptide-conjugated phosphodiamidate morpholino oligomer (P-PMO) Pip6a-PMO, for restoration of cardiac dystrophin and functional rescue in DMD mice- the mdx mouse and the less well characterised Cmah-/-mdx mouse (which carry a human-like mutation in the mouse Cmah gene as well as a mutation in DMD). In our first study male mdx mice were administered Pip6a-PMO, i.v, fortnightly from 12 to 30 weeks of age alongside mock-injected age-matched mdx and C57BL10 controls. Mice received 4 doses of 18 mg/kg followed by 8 doses of 12.5 mg/kg. The cardiac function of the mice was analysed 2 weeks after their final injection by MRI followed by conductance catheter and their muscles were harvested for dystrophin quantification. In the second study, male Cmah-/-mdx mice, received 12.5 mg/kg Pip6a-PMO, i.v fortnightly from 8 to 26 weeks and assessed by MRI at 3 time points (12, 18 and 28 weeks) alongside mock-injected age-matched mdx, C57BL10 and Cmah-/-mdx controls. The mice also underwent MEMRI and conductance catheter at 28 weeks. This allowed us to characterise the cardiac phenotype of Cmah-/-mdx mice as well as assess the effects of P-PMO on cardiac function. Pip6a-PMO treatment resulted in significant restoration of dystrophin in mdx and Cmah-/-mdx mice (37.5% and 51.6%, respectively), which was sufficient to significantly improve cardiac function, ameliorating both right and left ventricular dysfunction. Cmah-/-mdx mice showed an abnormal response to dobutamine stress test and this was completely ameliorated by PIP6a-PMO treatment. These encouraging data suggest that total restoration of dystrophin may not be required to significantly improve cardiac outcome in DMD patients and that it may be realistic to expect functional improvements with modest levels of dystrophin restoration which may be very achievable in future clinical trials.
- Published
- 2018
21. P7 Profile of circadianly regulated metabolic genes in dystrophic heart
- Author
-
K Meijboom, Wood Mja., Melissa Bowerman, Corinne A. Betts, Graham McClorey, and van Westering Tle.
- Subjects
medicine.medical_specialty ,biology ,business.industry ,Duchenne muscular dystrophy ,Cardiomyopathy ,Disease ,Carbohydrate metabolism ,medicine.disease ,Endocrinology ,Internal medicine ,Diabetes mellitus ,biology.protein ,medicine ,Circadian rhythm ,medicine.symptom ,Dystrophin ,business ,Wasting - Abstract
Duchenne muscular dystrophy (DMD) is a monogenic disorder caused by the lack of the integral structural protein, dystrophin, which results in severe muscle wasting and cardiomyopathy in affected boys. Indeed, cardiorespiratory complications are the predominant cause for mortality in DMD patients. We have recently shown that circadian rhythm is disrupted in dystrophic mice as a direct result of the lack of dystrophin protein. It is well reported that disruption of circadian rhythmicity leads to perturbed metabolism and an array of disorders including obesity, diabetes and cardiovascular disease. Disturbed cardiac metabolism in DMD patients and dystrophic mice is also well described, and thus it would be interesting to learn whether pertinent metabolic genes which are known to be circadianly regulated, are disrupted in dystrophic mice. Here we show for the first time, significant changes in the differential expression patterns of multiple genes involved in free fatty acid and glucose metabolism, in 2 mouse models of DMD compared to control mice. These findings provide the foundation for further research to better understand the metabolic/circadian milieu and its effect on dystrophic heart, so that we may devise strategies to augment cardiac metabolism, in an effort to halt the deterioration in cardiac phenotype.
- Published
- 2018
22. Peptide Nanoparticle Delivery of Charge-Neutral Splice-Switching Morpholino Oligonucleotides
- Author
-
Amer F. Saleh, Ülo Langel, Mattias Hällbrink, Michael J. Gait, Graham McClorey, Samir El Andaloussi, Suzan M. Hammond, Matthew J.A. Wood, C. I. Edvard Smith, Peter Järver, Andrey A. Arzumanov, and Eman M. Zaghloul
- Subjects
Morpholino ,Molecular Sequence Data ,Peptide ,Biology ,Biochemistry ,Morpholinos ,Muscular Atrophy, Spinal ,Mice ,Agammaglobulinemia ,Drug Discovery ,Genetics ,Animals ,Amino Acid Sequence ,Molecular Biology ,Cells, Cultured ,chemistry.chemical_classification ,Liposome ,Oligonucleotide ,RNA ,Genetic Diseases, X-Linked ,Biological activity ,Original Articles ,Transfection ,Muscular Dystrophy, Duchenne ,Disease Models, Animal ,chemistry ,Nucleic acid ,Nanoparticles ,Molecular Medicine ,Peptides - 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.
- Published
- 2015
23. Splice-Switching Therapy for Spinal Muscular Atrophy
- Author
-
Wood Mja., K Meijboom, and Graham McClorey
- Subjects
0301 basic medicine ,clinical trials ,lcsh:QH426-470 ,splice-switching oligonucleotides ,business.industry ,Mechanism (biology) ,Genetic disorder ,Spinal muscular atrophy ,Disease ,Review ,medicine.disease ,SMA ,Bioinformatics ,Clinical trial ,lcsh:Genetics ,03 medical and health sciences ,Exon ,030104 developmental biology ,Splice switching ,Genetics ,medicine ,business ,Genetics (clinical) ,spinal muscular atrophy - Abstract
Spinal muscular atrophy (SMA) is a genetic disorder with severity ranging from premature death in infants to restricted motor function in adult life. Despite the genetic cause of this disease being known for over twenty years, only recently has a therapy been approved to treat the most severe form of this disease. Here we discuss the genetic basis of SMA and the subsequent studies that led to the utilization of splice switching oligonucleotides to enhance production of SMN protein, which is absent in patients, through a mechanism of exon inclusion into the mature mRNA. Whilst approval of oligonucleotide-based therapies for SMA should be celebrated, we also discuss some of the limitations of this approach and alternate genetic strategies that are currently underway in clinical trials.
- Published
- 2017
24. Corrigendum: Identification of novel, therapy-responsive protein biomarkers in a mouse model of Duchenne muscular dystrophy by aptamer-based serum proteomics
- Author
-
Graham McClorey, Michael J. Gait, A. Blain, Corinne A. Betts, Thomas C. Roberts, Raquel Manzano, Hanns Lochmüller, Anna M.L. Coenen-Stass, Matthew J.A. Wood, and Amer F. Saleh
- Subjects
musculoskeletal diseases ,0301 basic medicine ,Male ,Proteomics ,congenital, hereditary, and neonatal diseases and abnormalities ,Serum proteomics ,Protein biomarkers ,Duchenne muscular dystrophy ,Aptamer ,Computational biology ,Protein Serine-Threonine Kinases ,Bioinformatics ,Article ,Biomarkers, Pharmacological ,Dystrophin ,03 medical and health sciences ,Mice ,medicine ,Animals ,Humans ,Multidisciplinary ,business.industry ,Gene Expression Profiling ,Blood Proteins ,Aptamers, Nucleotide ,Oligonucleotides, Antisense ,medicine.disease ,Corrigenda ,Phosphoric Monoester Hydrolases ,Mice, Inbred C57BL ,Muscular Dystrophy, Duchenne ,ADAM Proteins ,Disease Models, Animal ,030104 developmental biology ,Gene Expression Regulation ,Mice, Inbred mdx ,Identification (biology) ,ADAMTS5 Protein ,business ,Calcium-Calmodulin-Dependent Protein Kinase Type 2 ,Protein Kinases - 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
- Published
- 2016
25. Antisense pre-treatment increases gene therapy efficacy in dystrophic muscles
- Author
-
Graham McClorey, Amédée Mollard, Stéphanie Lorain, Sofia Benkhelifa-Ziyyat, Susan Jarmin, Thomas Voit, Anita Le Heron, Maëva Le Hir, Laura Julien, Matthew J.A. Wood, Cécile Peccate, George Dickson, HAL-UPMC, Gestionnaire, Institut de Myologie, Université Pierre et Marie Curie - Paris 6 (UPMC)-Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Association française contre les myopathies (AFM-Téléthon)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Handicap neuromusculaire : Physiopathologie, Biothérapie et Pharmacologies appliquées (END-ICAP), Université de Versailles Saint-Quentin-en-Yvelines (UVSQ)-Institut National de la Santé et de la Recherche Médicale (INSERM), South Parks Road, University of London [London], and Centre National de la Recherche Scientifique (CNRS)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Association française contre les myopathies (AFM-Téléthon)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Université Pierre et Marie Curie - Paris 6 (UPMC)
- Subjects
0301 basic medicine ,Morpholino ,Duchenne muscular dystrophy ,Genetic enhancement ,viruses ,Genetic Vectors ,[SDV.GEN] Life Sciences [q-bio]/Genetics ,Biology ,Pharmacology ,medicine.disease_cause ,Morpholinos ,Dystrophin ,03 medical and health sciences ,Sarcolemma ,Genetics ,medicine ,Myocyte ,Animals ,Humans ,Muscular dystrophy ,Muscle, Skeletal ,Molecular Biology ,Adeno-associated virus ,Genetics (clinical) ,[SDV.GEN]Life Sciences [q-bio]/Genetics ,Gene Transfer Techniques ,General Medicine ,Exons ,Genetic Therapy ,Dependovirus ,Muscular Dystrophy, Animal ,Oligonucleotides, Antisense ,medicine.disease ,Molecular biology ,3. Good health ,Muscular Dystrophy, Duchenne ,030104 developmental biology ,biology.protein ,Mice, Inbred mdx - Abstract
International audience; In preclinical models for Duchenne muscular dystrophy, dystrophin restoration during adeno-associated virus (AAV)-U7-mediated exon-skipping therapy was shown to decrease drastically after six months in treated muscles. This decline in efficacy is strongly correlated with the loss of the therapeutic AAV genomes, probably due to alterations of the dystrophic myofiber membranes. To improve the membrane integrity of the dystrophic myofibers at the time of AAV-U7 injection, mdx muscles were pre-treated with a single dose of the peptide-phosphorodiamidate morpholino (PPMO) antisense oligonucleotides that induced temporary dystrophin expression at the sarcolemma. The PPMO pre-treatment allowed efficient maintenance of AAV genomes in mdx muscles and enhanced the AAV-U7 therapy effect with a ten-fold increase of the protein level after 6 months. PPMO pre-treatment was also beneficial to AAV-mediated gene therapy with transfer of micro-dystrophin cDNA into muscles. Therefore, avoiding vector genome loss after AAV injection by PPMO pre-treatment would allow efficient long-term restoration of dystrophin and the use of lower and thus safer vector doses for Duchenne patients.
- Published
- 2016
26. Antisense oligonucleotide corrects splice abnormality in hereditary myopathy with lactic acidosis
- Author
-
Graham McClorey, Marina Toompuu, Petter Schandl Sanaker, and Laurence A. Bindoff
- Subjects
Iron-Sulfur Proteins ,Metabolic myopathy ,Exercise intolerance ,Biology ,Transfection ,Muscular Diseases ,Genetics ,medicine ,Humans ,splice ,Myopathy ,Cells, Cultured ,Skeletal muscle ,Genetic Therapy ,General Medicine ,Oligonucleotides, Antisense ,medicine.disease ,Molecular biology ,Alternative Splicing ,medicine.anatomical_structure ,Lactic acidosis ,RNA splicing ,biology.protein ,Cancer research ,Acidosis, Lactic ,ISCU ,medicine.symptom - Abstract
Hereditary myopathy with lactic acidosis (HML) (OMIM #255125) presents in childhood with exercise intolerance and muscle pain on trivial exercise, lactic acidosis, dyspnoea, palpitations, and rhabdomyolysis which can be fatal. The disease is recessively inherited and caused by a deep intronic, single base transition in the iron–sulfur cluster scaffold, ISCU gene that causes retention of a pseudoexon and introduction of a premature termination codon. IscU protein deficiency causes secondary defects in several iron–sulfur dependant proteins, including enzymes involved in aerobic energy metabolism. We have shown in a previous study that the splice abnormality affects skeletal muscle more than other tissues, leading to the purely muscular phenotype. Antisense oligonucleotides (AOs) have been able to redirect mRNA splicing in a number of disease models, and show promise in clinical studies. We designed 2′O-methyl phosphorothioate AOs targeting either splice site of the detrimental HML pseudoexon. The acceptor site AO effectively redirected splicing towards the normal state in cultured muscle fibroblasts, whilst the donor site AO promoted pseudoexon inclusion in both patient and control cells. Our results show that AO therapy seems feasible in HML, but care must be taken to avoid adverse splicing effects.
- Published
- 2012
27. DMD CLINICAL THERAPIES II
- Author
-
S Standley, I. Antonijevic, E. Purcell-Estabrook, P. Nadella, Z. Zhong, C. Shivalila, J Zhang, N Iwamoto, J. Davis, K. Longo, L. Guo, A. Durbin, F. Liu, G Lu, Carlo Rinaldi, M. Byrne, Graham McClorey, N Kothari, C Vargeese, and Matthew J.A. Wood
- Subjects
Neurology ,Pediatrics, Perinatology and Child Health ,Neurology (clinical) ,Genetics (clinical) - Published
- 2018
28. Antisense Oligonucleotide-induced Exon Skipping Across the Human Dystrophin Gene Transcript
- Author
-
Penny L Harding, Steve D. Wilton, Graham McClorey, Abbie M Fall, Catherine Coleman, and Sue Fletcher
- Subjects
musculoskeletal diseases ,congenital, hereditary, and neonatal diseases and abnormalities ,Transcription, Genetic ,Mature messenger RNA ,Duchenne muscular dystrophy ,Molecular Sequence Data ,Nonsense mutation ,Dystrophin ,03 medical and health sciences ,Exon ,0302 clinical medicine ,Utrophin ,Drug Discovery ,medicine ,Genetics ,Humans ,Molecular Biology ,Cells, Cultured ,030304 developmental biology ,Pharmacology ,0303 health sciences ,Base Sequence ,biology ,Exons ,Oligonucleotides, Antisense ,medicine.disease ,Exon skipping ,RNA splicing ,biology.protein ,Molecular Medicine ,030217 neurology & neurosurgery - Abstract
Protein-truncating mutations in the dystrophin gene lead to the most common childhood form of muscle wasting, Duchenne muscular dystrophy. Becker muscular dystrophy, a condition that typically arises from dystrophin gene lesions that do not disrupt the reading frame, clearly indicates that substantial domains of the dystrophin protein are not essential. Potential therapeutic intervention exists during pre-mRNA splicing, whereby selected exons are excised to either remove nonsense mutations or restore the reading frame around frame-shifting mutations from the mature mRNA. Appropriately designed antisense oligonucleotides (AOs), directed at amenable splicing motifs across the dystrophin gene transcript, block exon recognition and/or spliceosome assembly so that targeted exons are removed from the mature mRNA. We describe a panel of AOs designed to induce skipping of every exon within the human dystrophin gene transcript, with the exception of the first and last exons. Every exon targeted in vitro could be removed from the dystrophin mRNA, although some exons are more efficiently excluded than others. No single motif has emerged as a universal AO annealing site for redirection of dystrophin pre-mRNA processing, although the general trend is that the most efficient compounds are directed at motifs in the first half of the target exon.
- Published
- 2007
- Full Text
- View/download PDF
29. Identification of novel, therapy-responsive protein biomarkers in a mouse model of Duchenne muscular dystrophy by aptamer-based serum proteomics
- Author
-
Anna M. L. Coenen-Stass, Graham McClorey, Raquel Manzano, Corinne A. Betts, Alison Blain, Amer F. Saleh, Michael J. Gait, Hanns Lochmüller, Matthew J. A. Wood, and Thomas C. Roberts
- Published
- 2015
- Full Text
- View/download PDF
30. Self-Assembly into Nanoparticles Is Essential for Receptor Mediated Uptake of Therapeutic Antisense Oligonucleotides
- Author
-
John F. Morris, Samir El-Andaloussi, Mark A. Behlke, Aurélie Goyenvalle, Matthew J.A. Wood, Graham McClorey, Yoshitsugu Aoki, Amer F. Saleh, Leif Benner, Joel Z. Nordin, Anna M.L. Coenen-Stass, Liz O'Donovan, Christian J. Leumann, Taavi Lehto, Antonio Garcia-Guerra, Siamon Gordon, Michael J. Gait, Kariem Ezzat, Taeyoung Koo, and Branislav Dugovic
- Subjects
Morpholino ,Duchenne muscular dystrophy ,Bioengineering ,Micelle ,Article ,Cell Line ,chemistry.chemical_compound ,Mice ,540 Chemistry ,medicine ,Animals ,General Materials Science ,Micelles ,Mice, Knockout ,Base Sequence ,Oligonucleotide ,Mechanical Engineering ,Scavenger Receptors, Class A ,General Chemistry ,Receptor-mediated endocytosis ,Exons ,Genetic Therapy ,Oligonucleotides, Antisense ,Condensed Matter Physics ,medicine.disease ,Exon skipping ,Mice, Inbred C57BL ,Muscular Dystrophy, Duchenne ,chemistry ,Biochemistry ,Cell culture ,Biophysics ,570 Life sciences ,biology ,Nanoparticles ,DNA - 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 knock-out 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 co-localization 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.
- Published
- 2015
31. An overview of the clinical application of antisense oligonucleotides for RNA-targeting therapies
- Author
-
Matthew J.A. Wood and Graham McClorey
- Subjects
Pharmacology ,Oligonucleotide ,RNA Splicing ,RNA ,Gene Expression ,Biology ,Oligonucleotides, Antisense ,Bioinformatics ,Protein expression ,Drug Discovery ,Antisense oligonucleotides ,Animals ,Humans ,Rna targeting - Abstract
Despite the discovery more than two decades ago that antisense oligonucleotides (ASOs) could be used to modulate protein expression, there have been only two antisense drugs approved for clinical use till date. Despite this low success rate, the antisense field is undergoing resurgence due to the development of more potent and nuclease resistant chemistries, as well as nanoparticle delivery systems that enhance delivery to target tissues. In this review, we introduce the predominant therapeutic strategies in the antisense field whilst highlighting recent clinical findings that demonstrate the significant potential of these approaches for development of novel therapies in several diseases.
- Published
- 2015
32. Implications for Cardiac Function Following Rescue of the Dystrophic Diaphragm in a Mouse Model of Duchenne Muscular Dystrophy
- Author
-
Graham McClorey, Dominic J. Wells, Corinne A. Betts, S Muses, Matthew J.A. Wood, Caroline Godfrey, Kieran Clarke, Carolyn A. Carr, Suzan M. Hammond, Amer F. Saleh, Caroline Woffindale, Michael J. Gait, and K E Wells
- Subjects
Male ,Duchenne muscular dystrophy ,Gene Expression ,Morpholinos ,Dystrophin ,0302 clinical medicine ,Muscular dystrophy ,Respiratory system ,0303 health sciences ,Multidisciplinary ,Reverse Transcriptase Polymerase Chain Reaction ,Heart ,Anatomy ,musculoskeletal system ,Magnetic Resonance Imaging ,Diaphragm (structural system) ,NADPH Oxidase 4 ,Cardiology ,Atrial Natriuretic Factor ,Cardiac function curve ,musculoskeletal diseases ,medicine.medical_specialty ,congenital, hereditary, and neonatal diseases and abnormalities ,Blotting, Western ,Diaphragm ,Biology ,Article ,03 medical and health sciences ,Internal medicine ,Physical Conditioning, Animal ,medicine ,Respiratory muscle ,Animals ,Humans ,Amino Acid Sequence ,Protein Precursors ,Muscle, Skeletal ,030304 developmental biology ,NADPH Oxidases ,Natriuretic Peptide, C-Type ,medicine.disease ,Exon skipping ,Mice, Inbred C57BL ,Muscular Dystrophy, Duchenne ,Radiography ,Disease Models, Animal ,biology.protein ,Mice, Inbred mdx ,Peptides ,030217 neurology & neurosurgery - 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.
- Published
- 2015
33. Prevention of exercised induced cardiomyopathy following Pip-PMO treatment in dystrophic mdx mice
- Author
-
Kieran Clarke, Corinne A. Betts, Graham McClorey, Miguel A. Varela, Anna M.L. Coenen-Stass, Suzan M. Hammond, Michael J. Gait, Caroline Godfrey, Carolyn A. Carr, Amer F. Saleh, Matthew J.A. Wood, and Thomas C. Roberts
- Subjects
musculoskeletal diseases ,Cardiomyopathy, Dilated ,medicine.medical_specialty ,congenital, hereditary, and neonatal diseases and abnormalities ,Cardiac fibrosis ,Duchenne muscular dystrophy ,Cardiomyopathy ,Gene Expression ,Magnetic Resonance Imaging, Cine ,Article ,Morpholinos ,Dystrophin ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Internal medicine ,Physical Conditioning, Animal ,Medicine ,Animals ,Humans ,Muscular dystrophy ,030304 developmental biology ,0303 health sciences ,Multidisciplinary ,biology ,business.industry ,Myocardium ,Cardiac muscle ,Skeletal muscle ,Anatomy ,medicine.disease ,Fibrosis ,Exon skipping ,Muscular Dystrophy, Duchenne ,Disease Models, Animal ,medicine.anatomical_structure ,Endocrinology ,Phenotype ,biology.protein ,Mice, Inbred mdx ,business ,Cardiomyopathies ,030217 neurology & neurosurgery ,Biomarkers - 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.
- Published
- 2015
34. Antisense oligonucleotide-induced exon skipping restores dystrophin expression in vitro in a canine model of DMD
- Author
-
Sue Fletcher, Patrick L. Iversen, Graham McClorey, Hong M. Moulton, and Steve D. Wilton
- Subjects
musculoskeletal diseases ,congenital, hereditary, and neonatal diseases and abnormalities ,mdx mouse ,Duchenne muscular dystrophy ,Genetic enhancement ,Blotting, Western ,Gene Expression ,Transfection ,Dystrophin ,Exon ,Dogs ,Genetics ,medicine ,Animals ,Humans ,Muscular dystrophy ,Molecular Biology ,Cells, Cultured ,biology ,Reverse Transcriptase Polymerase Chain Reaction ,Alternative splicing ,Exons ,Genetic Therapy ,Oligonucleotides, Antisense ,medicine.disease ,Molecular biology ,Exon skipping ,Muscular Dystrophy, Duchenne ,Alternative Splicing ,biology.protein ,Molecular Medicine - Abstract
Manipulation of pre-mRNA splicing by antisense oligonucleotides (AOs) offers considerable potential for a number of genetic disorders. One of these is Duchenne muscular dystrophy (DMD), where mutations in the dystrophin gene typically result in premature termination of translation that causes a loss of functional protein. AOs can induce exon skipping such that the mutation is by-passed and the reading frame restored, producing an internally deleted protein similar to that found in the milder Becker muscular dystrophy. To date, this approach has been applied to the mdx mouse model in vitro and in vivo and in human myoblast cultures. Here, we report the application of AO-directed exon skipping to induce dystrophin expression in vitro in a canine model of DMD, golden retriever muscular dystrophy (GRMD). The efficacy of 2'-O-methyl phosphorothioate (2OMe), phosphorodiamidate morpholino oligomers (PMOs) and peptide-linked PMOs (PMO-Pep) to induce dystrophin expression was assessed. The 2OMe chemistry was only effective for short-term induction of corrected transcript and could not induce detectable dystrophin protein. The PMO chemistry generally induced limited exon skipping at only high concentrations; however, a low level of dystrophin protein was produced in treated cells. Use of the PMO-Pep, applied here for the first time to a DMD model, was able to induce high and sustained levels of exon skipping and induced the highest level of dystrophin expression with no apparent adverse effects upon the cells. The induction of dystrophin in the GRMD model offers the potential for further testing of AO delivery regimens in a larger animal model of DMD, in preparation for application in human clinical trials.
- Published
- 2006
35. Bi-specific splice-switching PMO oligonucleotides conjugated via a single peptide active in a mouse model of Duchenne muscular dystrophy
- Author
-
Graham McClorey, Michael J. Gait, Peter Järver, Fazel Shabanpoor, Amer F. Saleh, and Matthew J.A. Wood
- Subjects
mdx mouse ,Morpholino ,Cell Survival ,Duchenne muscular dystrophy ,Activin Receptors, Type II ,RNA Splicing ,Cell-Penetrating Peptides ,Biology ,Morpholinos ,Dystrophin ,Exon ,Mice ,Chemical Biology and Nucleic Acid Chemistry ,In vivo ,Genetics ,medicine ,Animals ,Cells, Cultured ,Oligonucleotide ,Exons ,medicine.disease ,Exon skipping ,Muscular Dystrophy, Duchenne ,Disease Models, Animal ,Biochemistry ,RNA splicing ,Mice, Inbred mdx - Abstract
The potential for therapeutic application of splice-switching oligonucleotides (SSOs) to modulate pre-mRNA splicing is increasingly evident in a number of diseases. However, the primary drawback of this approach is poor cell and in vivo oligonucleotide uptake efficacy. Biological activities can be significantly enhanced through the use of synthetically conjugated cationic cell penetrating peptides (CPPs). Studies to date have focused on the delivery of a single SSO conjugated to a CPP, but here we describe the conjugation of two phosphorodiamidate morpholino oligonucleotide (PMO) SSOs to a single CPP for simultaneous delivery and pre-mRNA targeting of two separate genes, exon 23 of the Dmd gene and exon 5 of the Acvr2b gene, in a mouse model of Duchenne muscular dystrophy. Conjugations of PMOs to a single CPP were carried out through an amide bond in one case and through a triazole linkage (‘click chemistry’) in the other. The most active bi-specific CPP–PMOs demonstrated comparable exon skipping levels for both pre-mRNA targets when compared to individual CPP–PMO conjugates both in cell culture and in vivo in the mdx mouse model. Thus, two SSOs with different target sequences conjugated to a single CPP are biologically effective and potentially suitable for future therapeutic exploitation.
- Published
- 2014
36. Improved antisense oligonucleotide induced exon skipping in themdx mouse model of muscular dystrophy
- Author
-
Graham McClorey, Sue Fletcher, Christopher J. Mann, Stephen D. Wilton, and K. Honeyman
- Subjects
musculoskeletal diseases ,congenital, hereditary, and neonatal diseases and abnormalities ,mdx mouse ,Duchenne muscular dystrophy ,Blotting, Western ,Nonsense mutation ,Muscular Dystrophies ,Dystrophin ,Mice ,Exon ,Drug Discovery ,Genetics ,medicine ,Animals ,Muscular dystrophy ,Molecular Biology ,Cells, Cultured ,Genetics (clinical) ,DNA Primers ,Messenger RNA ,Base Sequence ,biology ,Reverse Transcriptase Polymerase Chain Reaction ,Exons ,Oligonucleotides, Antisense ,medicine.disease ,Molecular biology ,Exon skipping ,Disease Models, Animal ,Mice, Inbred mdx ,biology.protein ,Molecular Medicine - Abstract
Background Duchenne muscular dystrophy (DMD) is a fatal genetic disorder caused by dystrophin gene mutations that preclude synthesis of a functional protein. One potential treatment of the disorder has utilised antisense oligoribonucleotides (AOs) to induce removal of disease-associated exons during pre-mRNA processing. Induced in-frame mRNA transcripts encode a shorter but functional dystrophin. We have investigated and improved the design of AOs capable of removing exon 23, and thus the disease-causing nonsense mutation, from mRNA in the mdx mouse model of DMD. Methods H−2Kb-tsA58 mdx cultures were transfected with complexes of Lipofectin and AOs. Exon skipping was detected by RT-PCR and subsequent protein production was demonstrated by Western blotting. AOs were delivered at a range of doses in order to compare relative efficiencies. Results We describe effective and reproducible exon 23 skipping with several AOs, including one as small as 17 nucleotides. Furthermore, the location of a sensitive exon 23 target site has been refined, whilst minimum effective doses have been estimated in vitro. These doses are significantly lower than previously reported and were associated with the synthesis of dystrophin protein in vitro. Conclusions These results demonstrate the increasing feasibility of an AO-based therapy for treatment of DMD. By refining AO design we have been able to reduce the size and the effective dose of the AOs and have dramatically improved the efficiency of the technique.
- Published
- 2002
37. WVE-210201, an investigational stereopure oligonucleotide therapy for Duchenne muscular dystrophy, induces Exon 51 skipping and dystrophin protein restoration
- Author
-
C Francis, A Zhong, J Zhang, M Frank-Kamenetsky, N Iwamoto, M Meena, D Butler, Graham McClorey, N Kothari, C Vargeese, Matthew J.A. Wood, S Mathieu, H Yang, Carlo Rinaldi, K Bowman, S Standley, M Shimizu, G Lu, and S Menon
- Subjects
biology ,Oligonucleotide ,business.industry ,Duchenne muscular dystrophy ,medicine.disease ,03 medical and health sciences ,Exon ,0302 clinical medicine ,Neurology ,030220 oncology & carcinogenesis ,Pediatrics, Perinatology and Child Health ,medicine ,Cancer research ,biology.protein ,Neurology (clinical) ,Dystrophin ,business ,030217 neurology & neurosurgery ,Genetics (clinical) - Published
- 2017
38. Design and application of bispecific splice-switching oligonucleotides
- Author
-
Samir El Andaloussi, U Tedebark, Suzan M. Hammond, Thomas C. Roberts, Moreno Pmd., Smith Cie., Graham McClorey, Wood Mja., and Burcu Bestas
- Subjects
Male ,RNA Splicing ,Computational biology ,Biology ,medicine.disease_cause ,Biochemistry ,Cell Line ,Dystrophin ,Exon ,Mice ,Splice switching ,Drug Discovery ,Genetics ,medicine ,Animals ,Humans ,Molecular Biology ,Mutation ,Base Sequence ,Oligonucleotide ,Translation (biology) ,Exons ,Muscular Dystrophy, Animal ,Myostatin ,Oligonucleotides, Antisense ,Mice, Inbred C57BL ,Muscular Dystrophy, Duchenne ,RNA splicing ,Mice, Inbred mdx ,Molecular Medicine ,Cleavable linker ,Linker ,Targeted Gene Repair - 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.
- Published
- 2014
39. Correlating In Vitro Splice Switching Activity With Systemic In Vivo Delivery Using Novel ZEN-modified Oligonucleotides
- Author
-
Yi-Huan Lee, Joel Z. Nordin, Sofia Stenler, Suzan M. Hammond, C. I. Edvard Smith, Kim A. Lennox, Caroline Godfrey, Samir El Andaloussi, Matthew J.A. Wood, Mark A. Behlke, Ashley M. Jacobi, Graham McClorey, and Miguel A. Varela
- Subjects
Duchenne muscular dystrophy ,ZEN modification ,03 medical and health sciences ,0302 clinical medicine ,In vivo ,Drug Discovery ,medicine ,030304 developmental biology ,Genetics ,splice switching ,0303 health sciences ,Nuclease ,biology ,Chemistry ,Oligonucleotide ,fungi ,lcsh:RM1-950 ,Transfection ,medicine.disease ,Exon skipping ,In vitro ,Cell biology ,lcsh:Therapeutics. Pharmacology ,030220 oncology & carcinogenesis ,biology.protein ,Molecular Medicine ,Original Article ,antisense oligonucleotides ,Dystrophin - 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.
- Published
- 2014
40. Cell-penetrating peptide–morpholino conjugates alter pre-mRNA splicing of DMD (Duchenne muscular dystrophy) and inhibit murine coronavirus replication in vivo
- Author
-
Hong M. Moulton, Bernard Lebleu, Sue Fletcher, Michael J. Buchmeier, Benjamin W. Neuman, Steve D. Wilton, Saïd Abes, David A. Stein, Graham McClorey, Patrick L. Iversen, Dynamique des interactions membranaires normales et pathologiques (DIMNP), and Centre National de la Recherche Scientifique (CNRS)-Université de Montpellier (UM)-Université Montpellier 2 - Sciences et Techniques (UM2)-Université Montpellier 1 (UM1)
- Subjects
Morpholino ,Morpholines ,RNA Splicing ,Duchenne muscular dystrophy ,Protein Sorting Signals ,Virus Replication ,medicine.disease_cause ,Antiviral Agents ,Biochemistry ,Dystrophin ,Mice ,03 medical and health sciences ,Drug Delivery Systems ,0302 clinical medicine ,In vivo ,RNA Precursors ,medicine ,Animals ,Humans ,[SDV.BBM]Life Sciences [q-bio]/Biochemistry, Molecular Biology ,Muscular dystrophy ,030304 developmental biology ,Coronavirus ,0303 health sciences ,biology ,Chemistry ,medicine.disease ,Molecular biology ,3. Good health ,Muscular Dystrophy, Duchenne ,Protein Transport ,030220 oncology & carcinogenesis ,biology.protein ,Cell-penetrating peptide ,Peptides ,Conjugate - Abstract
The cellular uptake of PMOs (phosphorodiamidate morpholino oligomers) can be enhanced by their conjugation to arginine-rich CPPs (cell-penetrating peptides). Here, we discuss our recent findings regarding (R-Ahx-R)4AhxB (Ahx is 6-aminohexanoic acid and B is β-alanine) CPP–PMO conjugates in DMD (Duchenne muscular dystrophy) and murine coronavirus research. An (R-Ahx-R)4AhxB–PMO conjugate was the most effective compound in inducing the correction of mutant dystrophin transcripts in myoblasts derived from a canine model of DMD. Similarly, normal levels of dystrophin expression were restored in the diaphragms of mdx mice, with treatment starting at the neonatal stage, and protein was still detecTable 22 weeks after the last dose of an (R-Ahx-R)4AhxB–PMO conjugate. Effects of length, linkage and carbohydrate modification of this CPP on the delivery of a PMO were investigated in a coronavirus mouse model. An (R-Ahx-R)4AhxB–PMO conjugate effectively inhibited viral replication, in comparison with other peptides conjugated to the same PMO. Shortening the CPP length, modifying it with a mannosylated serine moiety or replacing it with the R9F2 CPP significantly decreased the efficacy of the resulting PPMO (CPP–PMO conjugate). We attribute the success of this CPP to its stability in serum and its capacity to transport PMO to RNA targets in a manner superior to that of poly-arginine CPPs.
- Published
- 2007
41. Identification of novel therapy-responsive protein biomarkers for Duchenne muscular dystrophy by aptamer-based serum proteomics
- Author
-
Michael J. Gait, A. Blain, Thomas C. Roberts, Anna M.L. Coenen-Stass, Larry Gold, Corinne A. Betts, Graham McClorey, Matthew J.A. Wood, Amer F. Saleh, Raquel Manzano, and Hanns Lochmüller
- Subjects
Serum proteomics ,Protein biomarkers ,business.industry ,Duchenne muscular dystrophy ,Aptamer ,Computational biology ,medicine.disease ,Neurology ,Pediatrics, Perinatology and Child Health ,medicine ,Identification (biology) ,Neurology (clinical) ,business ,Genetics (clinical) - Published
- 2015
42. The physiological consequences of different levels of dystrophin following antisense based exon-skipping in the mdx mouse
- Author
-
S Muses, E O'Donovan, Matthew J.A. Wood, Caroline Godfrey, R L Terry, M J Gait, John C. W. Hildyard, Graham McClorey, K E Wells, S El Andaloussi, Ornella Cappellari, Suzan M. Hammond, Dominic J. Wells, Corinne A. Betts, and Thibault Coursindel
- Subjects
medicine.medical_specialty ,mdx mouse ,Specific force ,biology ,business.industry ,musculoskeletal system ,medicine.disease ,Exon skipping ,Pathophysiology ,Diaphragm (structural system) ,Exon ,Endocrinology ,Neurology ,Fibrosis ,Internal medicine ,Pediatrics, Perinatology and Child Health ,medicine ,biology.protein ,Neurology (clinical) ,business ,Dystrophin ,Genetics (clinical) - Abstract
We examined the effects on muscle physiology of restoring different levels of dystrophin in mdx mice with established dystrophic pathophysiology (12 weeks and older). Dystrophin expression was induced very efficiently using cell penetrating peptides linked to an antisense sequencing targeting exon 23 which contains a premature stop mutation. We assessed muscle physiology in the tibialis anterior (TA) muscle of the mouse using a terminally anaesthetised in situ protocol. To assess muscle physiology in the diaphragm we used strips of diaphragm in an in-vitro system. In both cases we examined the force–frequency relationship and established maximum specific tetanic force. We then subjected the muscles to a 10% stretch while stimulating them to contract. This eccentric exercise was highly damaging to dystrophic muscle. We present data showing that 15% of normal levels of dystrophin were sufficient to prevent eccentric exercise induced damage following a single dose of Pip6a-PMO. Chronic intravenous (IV) administration had a cumulative effect and we show that restoration of 50% of normal levels of dystrophin produced a 40% improvement in maximum specific force. Intraperitoneal administration of a single dose of B-PMO produced an 88% increase in maximum specific force as well as protecting against eccentric exercise induced damage in the diaphragm. Similar results were obtained in the diaphragm with chronic IV delivery of Pip6a-PMO at the same dose as the studies in the TA, even when treating older mice with extensive fibrosis in the diaphragm. While caution must be applied when extrapolating these results to DMD patients, the results suggest that moderate levels of dystrophin may be sufficient to slow-down or possibly prevent disease progression whereas higher levels of dystrophin will also improve muscle force production.
- Published
- 2015
43. Extracellular microRNAs are dynamic non-vesicular biomarkers of muscle turnover
- Author
-
Matthew J.A. Wood, Deborah Briggs, Ian L. Sargent, Chris Gardiner, Caroline Godfrey, Jennifer E. Morgan, Pieter Vader, Thomas C. Roberts, Yoshitsugu Aoki, and Graham McClorey
- Subjects
Male ,mdx mouse ,Duchenne muscular dystrophy ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Genetics ,medicine ,Extracellular ,Animals ,Regeneration ,Muscular dystrophy ,Muscle, Skeletal ,Myogenin ,030304 developmental biology ,0303 health sciences ,biology ,Skeletal muscle ,Blood Proteins ,medicine.disease ,Molecular biology ,Cell biology ,Mice, Inbred C57BL ,Muscular Dystrophy, Duchenne ,MicroRNAs ,medicine.anatomical_structure ,biology.protein ,Mice, Inbred mdx ,RNA ,Dystrophin ,ITGA7 ,030217 neurology & neurosurgery ,Biomarkers - Abstract
Extracellular microRNAs (miRNAs) are promising biomarkers of the inherited muscle wasting condition Duchenne muscular dystrophy, as they allow non-invasive monitoring of either disease progression or response to therapy. In this study, serum miRNA profiling reveals a distinct extracellular miRNA signature in dystrophin-deficient mdx mice, which shows profound dose-responsive restoration following dystrophin rescue. Extracellular dystrophy-associated miRNAs (dystromiRs) show dynamic patterns of expression that mirror the progression of muscle pathology in mdx mice. Expression of the myogenic miRNA, miR-206 and the myogenic transcription factor myogenin in the tibialis anterior muscle were found to positively correlate with serum dystromiR levels, suggesting that extracellular miRNAs are indicators of the regenerative status of the musculature. Similarly, extracellular dystromiRs were elevated following experimentally-induced skeletal muscle injury and regeneration in non-dystrophic mice. Only a minority of serum dystromiRs were found in extracellular vesicles, whereas the majority were protected from serum nucleases by association with protein/lipoprotein complexes. In conclusion, extracellular miRNAs are dynamic indices of pathophysiological processes in skeletal muscle.
- Published
- 2013
44. Expression analysis in multiple muscle groups and serum reveals complexity in the microRNA transcriptome of the mdx mouse with implications for therapy
- Author
-
Caroline Godfrey, Matthew J.A. Wood, Samir El Andaloussi, Corinne A. Betts, C. I. Edvard Smith, Graham McClorey, K. Emelie M. Blomberg, Michael J. Gait, Thibault Coursindel, and Thomas C. Roberts
- Subjects
Duchenne muscular dystrophy ,mdx mouse ,Morpholino ,microRNA ,lcsh:RM1-950 ,Biology ,medicine.disease ,Molecular biology ,Cell biology ,Transcriptome ,lcsh:Therapeutics. Pharmacology ,Drug Discovery ,Gene expression ,medicine ,biology.protein ,Molecular Medicine ,Gene silencing ,Original Article ,Dystrophin ,mdx ,microarray - Abstract
MicroRNAs (miRNAs) are a class of small RNAs that regulate gene expression and are implicated in wide-ranging cellular processes and pathological conditions including Duchenne muscular dystrophy (DMD). We have compared differential miRNA expression in proximal and distal limb muscles, diaphragm, heart and serum in the mdx mouse relative to wild-type controls. Global transcriptome analysis revealed muscle-specific patterns of differential miRNA expression as well as a number of changes common between tissues, including previously identified dystromirs. In the case of miR-31 and miR-34c, upregulation of primary-miRNA transcripts, precursor hairpins and all mature miRNAs derived from the same transcript or miRNA cluster, strongly suggests transcriptional regulation of these miRNAs. The most striking differences in differential miRNA expression were between muscle tissue and serum. Specifically, miR-1, miR-133a, and miR-206 were highly abundant in mdx serum but downregulated or modestly upregulated in muscle, suggesting that these miRNAs are promising disease biomarkers. Indeed, the relative serum levels of these miRNAs were normalized in response to peptide-phosphorodiamidate morpholino oligonucleotide (PMO) mediated dystrophin restoration therapy. This study has revealed further complexity in the miRNA transcriptome of the mdx mouse, an understanding of which will be valuable in the development of novel therapeutics and for monitoring their efficacy.
- Published
- 2013
45. Dual Myostatin and Dystrophin Exon Skipping by Morpholino Nucleic Acid Oligomers Conjugated to a Cell-penetrating Peptide Is a Promising Therapeutic Strategy for the Treatment of Duchenne Muscular Dystrophy
- Author
-
Amer F. Saleh, Linda Popplewell, Michael J. Gait, Matthew J.A. Wood, Graham McClorey, Alberto Malerba, George Dickson, and Jagjeet K Kang
- Subjects
Duchenne muscular dystrophy ,musculoskeletal diseases ,congenital, hereditary, and neonatal diseases and abnormalities ,medicine.medical_specialty ,Morpholino ,Myostatin ,dystrophin ,Internal medicine ,Drug Discovery ,medicine ,Soleus muscle ,biology ,lcsh:RM1-950 ,musculoskeletal system ,medicine.disease ,Molecular biology ,Exon skipping ,Muscle atrophy ,lcsh:Therapeutics. Pharmacology ,Endocrinology ,myostatin ,Sarcopenia ,biology.protein ,Molecular Medicine ,Original Article ,antisense oligonucleotides ,medicine.symptom ,Dystrophin ,exon skipping - Abstract
The knockdown of myostatin, a negative regulator of skeletal muscle mass may have important implications in disease conditions accompanied by muscle mass loss like cancer, HIV/AIDS, sarcopenia, muscle atrophy, and Duchenne muscular dystrophy (DMD). In DMD patients, where major muscle loss has occurred due to a lack of dystrophin, the therapeutic restoration of dystrophin expression alone in older patients may not be sufficient to restore the functionality of the muscles. We recently demonstrated that phosphorodiamidate morpholino oligomers (PMOs) can be used to re-direct myostatin splicing and promote the expression of an out-of-frame transcript so reducing the amount of the synthesized myostatin protein. Furthermore, the systemic administration of the same PMO conjugated to an octaguanidine moiety (Vivo-PMO) led to a significant increase in the mass of soleus muscle of treated mice. Here, we have further optimized the use of Vivo-PMO in normal mice and also tested the efficacy of the same PMO conjugated to an arginine-rich cell-penetrating peptide (B-PMO). Similar experiments conducted in mdx dystrophic mice showed that B-PMO targeting myostatin is able to significantly increase the tibialis anterior (TA) muscle weight and when coadministered with a B-PMO targeting the dystrophin exon 23, it does not have a detrimental interaction. This study confirms that myostatin knockdown by exon skipping is a potential therapeutic strategy to counteract muscle wasting conditions and dual myostatin and dystrophin skipping has potential as a therapy for DMD.Molecular Therapy - Nucleic Acids (2012) 1, e62; doi:10.1038/mtna.2012.54; published online 18 December 2012.
- Published
- 2012
- Full Text
- View/download PDF
46. Induced dystrophin exon skipping in human muscle explants
- Author
-
Steve D. Wilton, Abbie M Fall, John E.J. Rasko, Patrick L. Iversen, Graham McClorey, Sue Fletcher, Hong M. Moulton, and Monique M. Ryan
- Subjects
congenital, hereditary, and neonatal diseases and abnormalities ,mdx mouse ,Genetic enhancement ,Duchenne muscular dystrophy ,RNA Splicing ,DNA Mutational Analysis ,Dystrophin ,Exon ,Mice ,medicine ,RNA Precursors ,Animals ,Humans ,Genetic Predisposition to Disease ,Genetic Testing ,Muscular dystrophy ,Muscle, Skeletal ,Genetics (clinical) ,Cells, Cultured ,biology ,Exons ,Genetic Therapy ,Oligonucleotides, Antisense ,medicine.disease ,Molecular biology ,Exon skipping ,Muscular Dystrophy, Duchenne ,Neurology ,Pediatrics, Perinatology and Child Health ,RNA splicing ,Mutation ,biology.protein ,Mice, Inbred mdx ,Neurology (clinical) - Abstract
Antisense oligonucleotide (AO) manipulation of pre-mRNA splicing of the dystrophin gene is showing promise in overcoming Duchenne muscular dystrophy (DMD)-causing mutations. To date, this approach has been limited to studies using animal models or cultured human muscle cells, and evidence that AOs can induce exon skipping in human muscle has yet to be shown. In this study, we used different AO analogues to induce exon skipping in muscle explants derived from normal and DMD human tissue. We propose that inducing exon skipping in human muscle explants is closer to in vivo conditions than cells in monolayer cultures, and may minimize the numbers of participants in Phase I clinical studies to demonstrate proof of principle of exon skipping in human muscle.
- Published
- 2006
47. Splicing intervention for Duchenne muscular dystrophy
- Author
-
Sue Fletcher, Graham McClorey, and Stephen D. Wilton
- Subjects
musculoskeletal diseases ,Pharmacology ,Genetics ,congenital, hereditary, and neonatal diseases and abnormalities ,Duchenne muscular dystrophy ,RNA Splicing ,Genetic Therapy ,Biology ,Oligonucleotides, Antisense ,medicine.disease ,Phenotype ,Exon skipping ,Dystrophin ,Muscular Dystrophy, Duchenne ,Exon ,RNA, Small Nuclear ,Drug Discovery ,RNA splicing ,medicine ,biology.protein ,Animals ,Humans ,Muscular dystrophy ,Gene - Abstract
The manipulation of pre-mRNA to alter gene transcript splicing patterns offers considerable potential for many genetic disorders. In particular, the targeted removal of one or more exons from a gene transcript can skip over, or compensate for, disease-causing mutations. Duchenne muscular dystrophy (DMD), the most common and severe form of muscular dystrophy, is one such disorder that could benefit from this strategy. Splicing modulation can convert a DMD phenotype into the less severe allelic Becker-like phenotype. Recent studies using antisense oligonucleotide-targeted exon skipping to induce near normal dystrophin in vivo in animal models, and in vitro in DMD cell lines, highlight the promise of this approach. On the basis of these successes, human clinical trials could be realized in the near future.
- Published
- 2005
48. P1 Peptide-conjugated phosphodiamidate morpholino treatment in mdx mice: cardiac dystrophin restoration and function
- Author
-
Caroline Godfrey, E. Greally, Guy A. MacGowan, V. Straub, Steve Laval, Graham McClorey, A. Blain, Matthew J.A. Wood, and M J Gait
- Subjects
chemistry.chemical_classification ,Morpholino ,biology ,Peptide ,Conjugated system ,Cell biology ,Neurology ,chemistry ,Pediatrics, Perinatology and Child Health ,biology.protein ,Neurology (clinical) ,Dystrophin ,Genetics (clinical) ,Function (biology) - Published
- 2014
49. P04 Advancing the potential of peptide-PMO compounds in exon skipping therapy for DMD
- Author
-
Matthew J.A. Wood, M J Gait, S El Andaloussi, Suzan M. Hammond, Caroline Godfrey, Thibault Coursindel, Graham McClorey, and Corinne A. Betts
- Subjects
medicine.medical_specialty ,Neurology ,business.industry ,Family medicine ,Pediatrics, Perinatology and Child Health ,medicine ,Neurology (clinical) ,Neurosurgery ,business ,Genetics (clinical) ,Exon skipping ,Child health - Abstract
P03 What do MHCn and MHCd antibodies recognise? L. Feng, R. Phadke, F. Muntoni, C. Sewry. Dubowitz Neuromuscular Centre, Great Ormond Street Hospital and Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK; UCL Institute of Neurology, National Hospital for Neurology and Neurosurgery, London WC1N 3BG, UK; Centre for Inherited Neuromuscular Disorders, Robert Jones and Agnes Hunt Orthopaedic Hospital NHS Trust, Oswestry, SY10 7AG, UK
- Published
- 2012
50. Small RNA-Mediated Epigenetic Myostatin Silencing
- Author
-
Kevin V. Morris, Graham McClorey, Matthew J.A. Wood, Samir El Andaloussi, and Thomas C. Roberts
- Subjects
Small interfering RNA ,Small RNA ,medicine.drug_class ,muscle degeneration ,lcsh:RM1-950 ,Histone deacetylase inhibitor ,Myostatin ,Biology ,Molecular biology ,Chromatin ,Cell biology ,lcsh:Therapeutics. Pharmacology ,Trichostatin A ,myostatin ,siRNA ,Drug Discovery ,medicine ,biology.protein ,Molecular Medicine ,Gene silencing ,Original Article ,transcriptional gene silencing ,Epigenetics ,epigenetic ,medicine.drug - Abstract
Myostatin (Mstn) is a secreted growth factor that negatively regulates muscle mass and is therefore a potential pharmacological target for the treatment of muscle wasting disorders such as Duchenne muscular dystrophy. Here we describe a novel Mstn blockade approach in which small interfering RNAs (siRNAs) complementary to a promoter-associated transcript induce transcriptional gene silencing (TGS) in two differentiated mouse muscle cell lines. Silencing is sensitive to treatment with the histone deacetylase inhibitor trichostatin A, and the silent state chromatin mark H3K9me2 is enriched at the Mstn promoter following siRNA transfection, suggesting epigenetic remodeling underlies the silencing effect. These observations suggest that long-term epigenetic silencing may be feasible for Mstn and that TGS is a promising novel therapeutic strategy for the treatment of muscle wasting disorders.
- Published
- 2012
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.