42 results on '"Davies, Kay E."'
Search Results
2. Long-term clinical follow-up of a family with Becker muscular dystrophy associated with a large deletion in the DMD gene.
- Author
-
Davies, Kay E and Vogt, Julie
- Subjects
- *
BECKER muscular dystrophy , *FACIOSCAPULOHUMERAL muscular dystrophy , *DUCHENNE muscular dystrophy , *NEUROMUSCULAR diseases , *MUSCLE weakness , *GENETIC mutation - Abstract
• DMD is caused by DMD gene mutations that result in a lack of dystrophin protein. • BMD is also caused by DMD mutations, but symptoms are milder compared with DMD. • We present a case study of a patient with BMD and his affected relatives. • The patient could walk at age 61 years, despite 46% of his DMD gene being missing. • These findings informed minigene constructs, a promising therapy option for DMD. Duchenne muscular dystrophy is a neuromuscular disease caused by DMD gene mutations that result in an absence of functional dystrophin protein. Patients with Duchenne experience progressive muscle weakness, are typically wheelchair dependent by their early teens, and develop respiratory and cardiac complications that lead to death in their twenties or thirties. Becker muscular dystrophy is also caused by DMD gene mutations, but symptoms are less severe and progression is slower compared with Duchenne. We describe a case study of a patient with Becker muscular dystrophy who was still ambulant at age 61 years and had a milder phenotype than Duchenne, despite 46% of his DMD gene being missing. His affected relatives had similarly mild phenotypes and clinical courses. These data guided the understanding of the criticality of various regions of dystrophin and informed the development of micro-dystrophin constructs to compensate for the absence of functional dystrophin in Duchenne. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
3. Circadian profiling in two mouse models of lysosomal storage disorders; Niemann Pick type-C and Sandhoff disease
- Author
-
Richardson, Katie, Livieratos, Achilleas, Dumbill, Richard, Hughes, Steven, Ang, Gauri, Smith, David A., Morris, Lauren, Brown, Laurence A., Peirson, Stuart N., Platt, Frances M., Davies, Kay E., and Oliver, Peter L.
- Published
- 2016
- Full Text
- View/download PDF
4. Corrigendum to “Long-term clinical follow-up of a family with Becker muscular dystrophy associated with a large deletion in the DMD gene” Neuromuscular Disorders 39 (2024) 5–9
- Author
-
Davies, Kay E. and Vogt, Julie
- Published
- 2024
- Full Text
- View/download PDF
5. Muscular Dystrophy --Reason for optimism?
- Author
-
Burton, Edward A. and Davies, Kay E.
- Subjects
Muscular dystrophy -- Research ,Dystrophin -- Genetic aspects ,Gene mutations -- Analysis ,Genetic translation -- Physiological aspects ,Post-translational modification -- Physiological aspects ,Biological sciences - Abstract
Research advances made in the last decade and half in characterizing the mechanisms underlying the development of various forms of muscular dystrophy augurs well for optimism in managing and treating muscular dystrophy. Discovery of dystrophin and its molecular implications in Duchenne type and Becker muscular dystrophies are discussed.
- Published
- 2002
6. Utrophin-dystrophin-deficient mice as a model for Duchenne muscular dystrophy
- Author
-
Deconinck, Anne E., Rafael, Jill A., Skinner, Judith A., Brown, Susan C., Potter, Allyson C., Metzinger, Laurent, Watt, Diana J., Dickson, J. George, Tinsley, Jonathon M., and Davies, Kay E.
- Subjects
Dystrophin -- Research ,Mice -- Health aspects ,Duchenne muscular dystrophy -- Research ,Biological sciences - Abstract
The fatal muscle wasting disease Duchenne muscular dystrophy is caused by the lack of dystrophin in muscle membranes. Dystrophin deficiency, however, does not affect mice. A study on mice with dystrophin and dystrophin-related protein utrophin deficiency reveal ultrastructural neuromuscular and myotendinous junction abnormalities that causes premature death. Results indicate that dystrophin and utrophin have complementary functions in developmental muscular pathways.
- Published
- 1997
7. Spinal muscular atrophy at the crossroads of basic science and therapy
- Author
-
Sleigh, James N., Grice, Stuart J., Davies, Kay E., and Talbot, Kevin
- Published
- 2013
- Full Text
- View/download PDF
8. Is Good Housekeeping the Key to Motor Neuron Survival?
- Author
-
Talbot, Kevin and Davies, Kay E.
- Subjects
Neurons ,Biological sciences - Abstract
To link to full-text access for this article, visit this link: http://dx.doi.org/10.1016/j.cell.2008.05.002 Byline: Kevin Talbot (1), Kay E. Davies (1) Abstract: Spinal muscular atrophy (SMA) is caused by a drastic reduction in the ubiquitously expressed SMN protein, which is critical for the correct assembly of the snRNP complexes required for RNA splicing. However, it is unclear why loss of SMN and altered snRNP assembly only seem to affect motor neurons. Reporting in this issue, challenge prior assumptions about the housekeeping function of SMN and demonstrate that loss of SMN leads to highly tissue-specific effects on splicing. Author Affiliation: (1) MRC Functional Genetics Unit, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, UK
- Published
- 2008
9. Treating Muscular Dystrophy with Stem Cells?
- Author
-
Davies, Kay E. and Grounds, Miranda D.
- Subjects
Stem cells ,Biological sciences - Abstract
To link to full-text access for this article, visit this link: http://dx.doi.org/10.1016/j.cell.2006.12.010 Byline: Kay E. Davies (1), Miranda D. Grounds (2) Abstract: There is currently no effective treatment for the devastating muscle-wasting disease Duchenne muscular dystrophy (DMD). Cossu and colleagues report in a recent Nature paper that transplantation of mesoangioblast stem cells may hold promise for treating DMD. Further studies are required to fully evaluate the clinical potential of these blood-vessel-associated stem cells. Author Affiliation: (1) MRC Functional Genetics Unit, University of Oxford, Oxford, UK (2) School of Anatomy & Human Biology, the University of Western Australia, Australia
- Published
- 2006
10. Disruption of SMN function by ectopic expression of the human SMN gene in Drosophila
- Author
-
Miguel-Aliaga, Irene, Chan, Yick Bun, Davies, Kay E., and van den Heuvel, Marcel
- Published
- 2000
- Full Text
- View/download PDF
11. Pharmacological advances for treatment in Duchenne muscular dystrophy.
- Author
-
Guiraud, Simon and Davies, Kay E
- Subjects
- *
TREATMENT of Duchenne muscular dystrophy , *DRUG development , *DRUG approval , *DISEASE progression , *CLINICAL drug trials - Abstract
Duchenne muscular dystrophy (DMD) is a lethal, X-linked muscle-wasting disease caused by lack of dystrophin, essential for muscle fibre integrity. Despite extensive pre-clinical studies, development of an effective treatment has proved challenging. More recently, significant progress has been made with the first drug approval using a genetic approach and the application of pharmacological agents which slow the progression of the disease. Drug development for DMD has mainly used two strategies: (1) the restoration of dystrophin expression or the expression of the compensatory utrophin protein as an efficient surrogate, and (2) the mitigation of secondary downstream pathological mechanisms. This review details current most promising pharmacological approaches and clinical trials aiming to tackle the pathogenesis of this multifaceted disorder. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
12. The antioxidant protein Oxr1 influences aspects of mitochondrial morphology.
- Author
-
Wu, Yixing, Davies, Kay E., and Oliver, Peter L.
- Subjects
- *
THERAPEUTIC use of antioxidants , *MITOCHONDRIAL pathology , *OXIDATIVE stress , *TREATMENT of neurodegeneration , *GENETIC overexpression , *LABORATORY mice - Abstract
Oxidative stress (OS) and mitochondrial dysfunction are implicated in neurodegenerative disease, suggesting that antioxidant defence systems are critical for cell survival in the central nervous system (CNS). Oxidation resistance 1 (OXR1) can protect against OS in cellular and mouse models of amyotrophic lateral sclerosis (ALS) when over-expressed, whereas deletion of Oxr1 in mice causes neurodegeneration. OXR1 has emerged therefore as an essential antioxidant protein that controls the susceptibility of neurons to OS. It has been suggested that OXR1 is localised to mitochondria, yet the functional significance of this has not been investigated in the context of neuronal cell death. In order to characterise the role of Oxr1 in mitochondria, we investigated its sub-mitochondrial localisation and demonstrate that specific isoforms are associated with the outer mitochondrial membrane, while the full-length Oxr1 protein is predominately cytoplasmic. Interestingly, cytoplamsic over-expression of these mitochondrially-localised isoforms was still able to protect against OS-induced cell death and prevent rotenone-induced mitochondrial morphological changes. To study the consequences of Oxr1 deletion in vivo , we utilised the bella ataxic mouse mutant. We were unable to identify defects in mitochondrial metabolism in primary cerebellar granule cells (GCs) from bella mice, however a reduction in mitochondrial length was observed in mutant GCs compared to those from wild-type. Furthermore, screening a panel of proteins that regulate mitochondrial morphology in bella GCs revealed de-regulation of phospho-Drp1(Ser616), a key mitochondrial fission regulatory factor. Our data provide new insights into the function of Oxr1, revealing that specific isoforms of this novel antioxidant protein are associated with mitochondria and that the modulation of mitochondrial morphology may be an important feature of its protective function. These results have important implications for the potential use of OXR1 in future antioxidant therapies. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
13. Microarray analysis of mdx mice expressing high levels of utrophin: Therapeutic implications for dystrophin deficiency
- Author
-
Baban, Dilair and Davies, Kay E.
- Subjects
- *
NEUROMUSCULAR diseases , *MEMBRANE proteins , *DYSTROPHIN , *GENE expression - Abstract
Abstract: Duchenne Muscular Dystrophy (DMD) is a fatal muscle wasting disorder caused by dystrophin deficiency. Previous work suggested that increased expression of the dystrophin-related protein utrophin in the mdx mouse can reduce the dystrophic pathophysiology. Physiological tests showed that the transgenic mouse muscle functioned in a way similar to normal muscle. More recently, it has become possible to analyse disease pathways using microarrays, a sensitive method to evaluate the efficacy of a therapeutic approach. We thus examined the gene expression profile of mdx mouse muscle compared to wild-type mouse muscle and compared the data with that obtained from the transgenic line overexpressing utrophin. The data confirm that the expression of utrophin in the mdx mouse muscle results in a global gene expression profile more similar to that seen for the wild-type mouse. This study confirms that a strategy to up-regulate utrophin is likely to be beneficial in dystrophin deficiency. [Copyright &y& Elsevier]
- Published
- 2008
- Full Text
- View/download PDF
14. Muscular dystrophy: from gene to patient
- Author
-
Hopkins, James C.A, Bia, Britta L, Crilley, Jenifer G, Boehm, Ernest A, Elizabeth Sang, A, Tinsley, Jonathon M, King, Linda M, Radda, George K, Davies, Kay E, and Clarke, Kieran
- Published
- 2000
15. The allure of stem cell therapy for muscular dystrophy
- Author
-
Grounds, Miranda D. and Davies, Kay E.
- Subjects
- *
DYSTROPHY , *NEUROMUSCULAR diseases , *MUSCULAR dystrophy in children , *MEMBRANE proteins - Abstract
Abstract: Duchenne muscular dystrophy (DMD) is a lethal muscle disease for which an effective treatment is urgently needed. The use of stem cells to produce normal muscle cells to replace the missing dystrophin protein has attracted much attention. Claims of success using stem cell treatment in animal models of human muscle diseases require careful evaluation and are not necessarily easily extrapolated to the clinical situation. Recent studies in the dystrophic dog model have been claimed to show that injected mesangioblasts, stem cells derived from blood vessels, reduce the severity of the disease. However, the authors’ interpretation of the results did not consider that benefits might arise from the concomitant use of immunosuppressive drugs alone. [Copyright &y& Elsevier]
- Published
- 2007
- Full Text
- View/download PDF
16. The role of utrophin in the potential therapy of Duchenne muscular dystrophy
- Author
-
Perkins, Kelly J. and Davies, Kay E.
- Subjects
- *
DYSTROPHIN , *THERAPEUTICS - Abstract
Duchenne muscular dystrophy is an X-linked recessive muscle wasting disease caused by the absence of the muscle cytoskeletal protein, dystrophin. Dystrophin is a member of the spectrin superfamily of proteins and is closely related in sequence similarity and functional motifs to three proteins that constitute the dystrophin related protein family, including the autosomal homologue, utrophin. An alternative strategy circumventing many problems associated with somatic gene therapies for Duchenne muscular dystrophy has arisen from the demonstration that utrophin can functionally substitute for dystrophin and its over-expression in muscles of dystrophin-null transgenic mice completely prevents the phenotype arising from dystrophin deficiency. One potential approach to increase utrophin levels in muscle for possible therapeutic purpose in humans is to increase expression of the utrophin gene at a transcriptional level via promoter activation. This has lead to an interest in the identification and manipulation of important regulatory regions and/or molecules that increase the expression of utrophin and their delivery to dystrophin-deficient tissue. As pre-existing cellular mechanisms are utilized, this approach would avoid many problems associated with conventional gene therapies. [Copyright &y& Elsevier]
- Published
- 2002
- Full Text
- View/download PDF
17. Rescue of a single yeast artificial chromosome from a cotransformation event utilizing segregation at meiosis
- Author
-
Németh, Andrea H., Hunter, Neil, Coleman, Michael P., Borts, Rhona H., Louis, Edward J., and Davies, Kay E.
- Published
- 1993
- Full Text
- View/download PDF
18. The fragile X syndrome
- Author
-
Flannery, Angela V., Hirst, Mark C., Knight, Samantha J.L., Ritchie, Rachael J., and Davies, Kay E.
- Published
- 1995
- Full Text
- View/download PDF
19. Exclusion of the gene responsible for facioscapulohumeral muscular dystrophy (FSH) at 6q23-q27
- Author
-
Passos-Bueno, M.Rita, Byth, Barbara, Love, Donald, Terwilliger, Joseph, Ott, Jurg, Rapaport, Debora, Vainzof, Mariz, Zatz, Mayana, and Davies, Kay E.
- Published
- 1991
- Full Text
- View/download PDF
20. Control of RNA transcription in nuclei and nucleoli of Physarum polycephalum
- Author
-
Davies, Kay E. and Walker, Ian O.
- Published
- 1978
- Full Text
- View/download PDF
21. Behavioural characterisation of the robotic mouse mutant
- Author
-
Oliver, Peter L., Keays, David A., and Davies, Kay E.
- Subjects
- *
ROBOTICS , *MOTOR ability , *PURKINJE cells , *LABORATORY mice - Abstract
Abstract: The ataxic mouse mutant robotic is characterised by progressive adult-onset Purkinje cell loss that occurs in a distinctive region-specific pattern. We report the first behavioural characterisation of this mutant and quantify its performance on tests of motor function, locomotor and exploratory activity over a time course that reflects specific stages of cell loss in the cerebellum. Robotic mutants are significantly impaired on the rotarod and static rod tests of coordination and their performance declined during aging. In addition, gait analysis revealed an increase in the severity of the ataxia displayed by mutants over time. Interestingly, spontaneous alternation testing in a T-maze was not significantly affected in robotic mice, unlike other ataxic mutants with more rapid and extensive cerebellar degeneration; robotic therefore provides an opportunity to investigate the necessity of specific Purkinje cell populations for various behavioural tasks. [Copyright &y& Elsevier]
- Published
- 2007
- Full Text
- View/download PDF
22. Identification and Characterization of Murine SCARA5, a Novel Class A Scavenger Receptor That Is Expressed by Populations of Epithelial Cells.
- Author
-
Yanyan Jiang, Oliver, Peter, Davies, Kay E., and Platt, Nick
- Subjects
- *
EPITHELIAL cells , *MICROORGANISMS , *IMMUNE response , *MACROPHAGES , *ESCHERICHIA coli , *STAPHYLOCOCCUS aureus - Abstract
Epithelia are positioned at a critical interface to prevent invasion by microorganisms from the environment. Pattern recognition receptors are important components of innate immunity because of their ability to interact with specific microbe-associated structures and initiate immune responses. Several distinct groups of receptors have been recognized. One of these, the scavenger receptors, has been classified into at least eight separate classes. The class A scavenger receptors are characterized by the presence of a collagen-like domain and include macrophage scavenger receptor type A (SR-A1 I/II, SCARA1) and MARCO (SCARA2). These receptors are known to make important contributions to host defense. Here, we identify a novel murine scavenger receptor, SCARA5, which has a structure typical of this class. The cDNA encodes 491 amino acids, which predict a type II protein that contains C-terminal intracellular, transmembrane, extracellular spacer, collagenous, and N-terminal scavenger receptor cysteine rich domains. Expression in Chinese hamster ovary cells confirmed that the receptor assembles as a homotrimer and is expressed at the plasma membrane. SCARA5-transfected cells bound Escherichia coli and Staphylococcus aureus, but not zymosan, in a polyanionic-inhibitable manner. Unlike other class A scavenger receptors, the receptor was unable to endocytose acetylated or oxidized low density lipoprotein. Quantitative RT-PCR and in situ hybridization demonstrate SCARA5 has a tissue and cellular distribution unique among class A scavenger receptors. Because of the restriction of SCARA5 transcripts to populations of epithelial cells, we propose that this receptor may play important roles in the innate immune activities of these cells. [ABSTRACT FROM AUTHOR]
- Published
- 2006
- Full Text
- View/download PDF
23. A-utrophin up-regulation in mdx skeletal muscle is independent of regeneration
- Author
-
Weir, Andrew P., Morgan, Jennifer E., and Davies, Kay E.
- Subjects
- *
DUCHENNE muscular dystrophy , *JUVENILE diseases , *DYSTROPHIN , *GENETIC mutation - Abstract
Duchenne muscular dystrophy is a fatal childhood disease caused by mutations that abolish the expression of dystrophin in muscle. Utrophin is a paralogue of dystrophin and can functionally replace it in skeletal muscle. A method to induce utrophin up-regulation in muscle should therefore be therapeutically useful in Duchenne muscular dystrophy. The search for such a method needs to be informed by an understanding of the mechanisms controlling utrophin expression in muscle. Two full length utrophin isoforms are expressed: A and B. A-utrophin is up-regulated in dystrophin deficient skeletal muscle and we sought to test the hypothesis that this up-regulation occurs as a consequence of ongoing regeneration. We measured utrophin expression by immunohistochemistry and immunoblotting in the oesophageal outer muscular layer and in γ-irradiated limb muscle from mdx mice. Skeletal muscle in these tissues is dystrophin deficient but not regenerating; we found that A-utrophin up-regulation still occurred. We conclude that utrophin up-regulation in skeletal muscle does not depend on regeneration. An alternative hypothesis involving competition for binding sites between utrophin and dystrophin is discussed. These results have important implications for future studies aiming to effect therapeutic utrophin up-regulation in Duchenne muscular dystrophy patients. [Copyright &y& Elsevier]
- Published
- 2004
- Full Text
- View/download PDF
24. The Evolutionarily Conserved Tre2/Bub2/Cdc16 (TBC), Lysin Motif (LysM), Domain Catalytic (TLDc) Domain Is Neuroprotective against Oxidative Stress.
- Author
-
Finelli, Mattéa J., Sanchez-Pulido, Luis, Liu, Kevin X., Davies, Kay E., and Oliver, Peter L.
- Subjects
- *
LYSINE , *NEUROPROTECTIVE agents , *OXIDATIVE stress , *THERMAL barrier coatings , *THERMOLUMINESCENCE dosimetry , *NEUROLOGICAL disorders , *NEURODEGENERATION , *GENETIC overexpression - Abstract
Oxidative stress is a pathological feature of many neurological disorders; therefore, utilizing proteins that are protective against such cellular insults is a potentially valuable therapeutic approach. Oxidation resistance 1 (OXR1) has been shown previously to be critical for oxidative stress resistance in neuronal cells; deletion of this gene causes neurodegeneration in mice, yet conversely, overexpression of OXR1 is protective in cellular and mouse models of amyotrophic lateral sclerosis. However, the molecular mechanisms involved are unclear. OXR1 contains the Tre2/Bub2/Cdc16 (TBC), lysin motif (LysM), domain catalytic (TLDc) domain, a motif present in a family of proteins including TBC1 domain family member 24 (TBC1D24), a protein mutated in a range of disorders characterized by seizures, hearing loss, and neurodegeneration. The TLDc domain is highly conserved across species, although the structure-function relationship is unknown. To understand the role of this domain in the stress response, we carried out systematic analysis of all mammalian TLDc domain-containing proteins, investigating their expression and neuroprotective properties in parallel. In addition, we performed a detailed structural and functional study of this domain in which we identified key residues required for its activity. Finally, we present a new mouse insertional mutant of Oxr1, confirming that specific disruption of the TLDc domain in vivo is sufficient to cause neurodegeneration. Our data demonstrate that the integrity of the TLDc domain is essential for conferring neuroprotection, an important step in understanding the functional significance of all TLDc domain-containing proteins in the cellular stress response and disease. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
25. Structure-activity relationships of 2-pyrimidinecarbohydrazides as utrophin modulators for the potential treatment of Duchenne muscular dystrophy.
- Author
-
Chatzopoulou, Maria, Conole, Daniel, Emer, Enrico, Rowley, Jessica A., Willis, Nicky J., Squire, Sarah E., Gill, Becky, Brough, Steve, Wilson, Francis X., Wynne, Graham M., Davies, Stephen G., Davies, Kay E., and Russell, Angela J.
- Subjects
- *
DUCHENNE muscular dystrophy , *LIPOPHILICITY , *STRUCTURE-activity relationships , *THERAPEUTICS - Abstract
[Display omitted] A therapeutic approach that holds the potential to treat all Duchenne muscular dystrophy (DMD) patient populations is utrophin modulation. Ezutromid, a first generation utrophin modulator which was later found to act via antagonism of the arylhydrocarbon receptor, progressed to Phase 2 clinical trials. Although interim data showed target engagement and functional improvements, ezutromid ultimately failed to meet its clinical endpoints. We recently described the identification of a new class of hydrazide utrophin modulators which has a different mechanism of action to ezutromid. In this study we report our early optimisation studies on this hydrazide series. The new analogues had significantly improved potency in cell-based assays, increased sp3 character and reduced lipophilicity, which also improved their physicochemical properties. A representative new analogue combining these attributes increased utrophin protein in dystrophic mouse cells showing it can be used as a chemical tool to reveal new insights regarding utrophin upregulation as a strategy for DMD therapeutic intervention. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
26. A Novel Mouse Model of a Patient Mucolipidosis II Mutation Recapitulates Disease Pathology.
- Author
-
Paton, Leigh, Bitoun, Emmanuelle, Kenyon, Janet, Priestman, David A., Oliver, Peter L., Edwards, Benjamin, Platt, Frances M., and Davies, Kay E.
- Subjects
- *
LIPIDOSES , *GLUCOSAMINE , *PHOSPHOTRANSFERASES , *MANNOSE 6-phosphate , *LYSOSOMES - Abstract
Mucolipidosis II (MLII) is a lysosomal storage disorder caused by loss of N-acetylglucosamine-1-phosphotransferase, which tags lysosomal enzymes with a mannose 6-phosphate marker for transport to the lysosome. In MLII, the loss of this marker leads to deficiency of multiple enzymes and non-enzymatic proteins in the lysosome, leading to the storage of multiple substrates. Here we present a novel mouse model of MLII homozygous for a patient mutation in the GNPTAB gene. Whereas the current gene knock-out mouse model of MLII lacks some of the characteristic features of the human disease, our novel mouse model more fully recapitulates the human pathology, showing growth retardation, skeletal and facial abnormalities, increased circulating lysosomal enzymatic activities, intracellular lysosomal storage, and reduced life span. Importantly, MLII behavioral deficits are characterized for the first time, including impaired motor function and psychomotor retardation. Histological analysis of the brain revealed progressive neurodegeneration in the cerebellum with severe Purkinje cell loss as the underlying cause of the ataxic gait. In addition, based on the loss of Npc2 (Niemann-Pick type C 2) protein expression in the brain, the mice were treated with 2-hydroxypropyl-β-cyclodextrin, a drug previously reported to rescue Purkinje cell death in a mouse model of Niemann-Pick type C disease. No improvement in brain pathology was observed. This indicates that cerebellar degeneration is not primarily triggered by loss of Npc2 function. This study emphasizes the value of modeling MLII patient mutations to generate clinically relevant mouse mutants to elucidate the pathogenic molecular pathways of MLII and address their amenability to therapy. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
27. Overexpression of survival motor neuron improves neuromuscular function and motor neuron survival in mutant SOD1 mice.
- Author
-
Turner, Bradley J., Alfazema, Neza, Sheean, Rebecca K., Sleigh, James N., Davies, Kay E., Horne, Malcolm K., and Talbot, Kevin
- Subjects
- *
MOTOR neurons , *MYONEURAL junction , *SPINAL muscular atrophy , *GENETICS of amyotrophic lateral sclerosis , *SUPEROXIDE dismutase genetics , *LABORATORY mice , *GENE expression - Abstract
Abstract: Spinal muscular atrophy results from diminished levels of survival motor neuron (SMN) protein in spinal motor neurons. Low levels of SMN also occur in models of amyotrophic lateral sclerosis (ALS) caused by mutant superoxide dismutase 1 (SOD1) and genetic reduction of SMN levels exacerbates the phenotype of transgenic SOD1G93A mice. Here, we demonstrate that SMN protein is significantly reduced in the spinal cords of patients with sporadic ALS. To test the potential of SMN as a modifier of ALS, we overexpressed SMN in 2 different strains of SOD1G93A mice. Neuronal overexpression of SMN significantly preserved locomotor function, rescued motor neurons, and attenuated astrogliosis in spinal cords of SOD1G93A mice. Despite this, survival was not prolonged, most likely resulting from SMN mislocalization and depletion of gems in motor neurons of symptomatic mice. Our results reveal that SMN upregulation slows locomotor deficit onset and motor neuron loss in this mouse model of ALS. However, disruption of SMN nuclear complexes by high levels of mutant SOD1, even in the presence of SMN overexpression, might limit its survival promoting effects in this specific mouse model. Studies in emerging mouse models of ALS are therefore warranted to further explore the potential of SMN as a modifier of ALS. [Copyright &y& Elsevier]
- Published
- 2014
- Full Text
- View/download PDF
28. AAV Genome Loss From Dystrophic Mouse Muscles During AAV-U7 snRNA-mediated Exon-skipping Therapy.
- Author
-
Le Hir, Maëva, Goyenvalle, Aurélie, Peccate, Cécile, Précigout, Guillaume, Davies, Kay E, Voit, Thomas, Garcia, Luis, and Lorain, Stéphanie
- Subjects
- *
LABORATORY mice , *EXONS (Genetics) , *ADENO-associated virus , *GENOMES , *CLINICAL trials - Abstract
In the context of future adeno-associated viral (AAV)-based clinical trials for Duchenne myopathy, AAV genome fate in dystrophic muscles is of importance considering the viral capsid immunogenicity that prohibits recurring treatments. We showed that AAV genomes encoding non-therapeutic U7 were lost from mdx dystrophic muscles within 3 weeks after intramuscular injection. In contrast, AAV genomes encoding U7ex23 restoring expression of a slightly shortened dystrophin were maintained endorsing that the arrest of the dystrophic process is crucial for maintaining viral genomes in transduced fibers. Indeed, muscles treated with low doses of AAV-U7ex23, resulting in sub-optimal exon skipping, displayed much lower titers of viral genomes, showing that sub-optimal dystrophin restoration does not prevent AAV genome loss. We also followed therapeutic viral genomes in severe dystrophic dKO mice over time after systemic treatment with scAAV9-U7ex23. Dystrophin restoration decreased significantly between 3 and 12 months in various skeletal muscles, which was correlated with important viral genome loss, except in the heart. Altogether, these data show that the success of future AAV-U7 therapy for Duchenne patients would require optimal doses of AAV-U7 to induce substantial levels of dystrophin to stabilize the treated fibers and maintain the long lasting effect of the treatment. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
29. Engineering Multiple U7snRNA Constructs to Induce Single and Multiexon-skipping for Duchenne Muscular Dystrophy.
- Author
-
Goyenvalle, Aurélie, Wright, Jordan, Babbs, Arran, Wilkins, Vivienne, Garcia, Luis, and Davies, Kay E
- Subjects
- *
MUSCULAR dystrophy , *DYSTROPHIN genes , *DYSTROPHIN , *MYOBLASTS , *DUCHENNE muscular dystrophy , *NEUROMUSCULAR diseases - Abstract
Duchenne muscular dystrophy (DMD) is a fatal muscle wasting disorder caused by mutations in the dystrophin gene. Antisense-mediated exon skipping is one of the most promising approaches for the treatment of DMD but still faces personalized medicine challenges as different mutations found in DMD patients require skipping of different exons. However, 70% of DMD patients harbor dystrophin gene deletions in a mutation-rich area or 'hot-spot' in the central genomic region. In this study, we have developed 11 different U7 small-nuclear RNA, to shuttle antisense sequences designed to mask key elements involved in the splicing of exons 45 to 55. We demonstrate that these constructs induce efficient exon skipping both in vitro in DMD patients' myoblasts and in vivo in human DMD (hDMD) mice and that they can be combined into a single vector to achieve a multi skipping of at least 3 exons. These very encouraging results provide proof of principle that efficient multiexon-skipping can be achieved using adeno-associated viral (AAV) vectors encoding multiple U7 small-nuclear RNAs (U7snRNAs), offering therefore very promising tools for clinical treatment of DMD. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
30. Current Status of Pharmaceutical and Genetic Therapeutic Approaches to Treat DMD.
- Author
-
Pichavant, Christophe, Aartsma-Rus, Annemieke, Clemens, Paula R, Davies, Kay E, Dickson, George, Takeda, Shin'ichi, Wilton, Steve D, Wolff, Jon A, Wooddell, Christine I, Xiao, Xiao, and Tremblay, Jacques P
- Subjects
- *
TREATMENT of Duchenne muscular dystrophy , *DYSTROPHIN , *ANTISENSE DNA , *GENE therapy , *DEGENERATION (Pathology) , *GENETIC code , *EXONS (Genetics) - Abstract
Duchenne muscular dystrophy (DMD) is a genetic disease affecting about one in every 3,500 boys. This X-linked pathology is due to the absence of dystrophin in muscle fibers. This lack of dystrophin leads to the progressive muscle degeneration that is often responsible for the death of the DMD patients during the third decade of their life. There are currently no curative treatments for this disease but different therapeutic approaches are being studied. Gene therapy consists of introducing a transgene coding for full-length or a truncated version of dystrophin complementary DNA (cDNA) in muscles, whereas pharmaceutical therapy includes the use of chemical/biochemical substances to restore dystrophin expression or alleviate the DMD phenotype. Over the past years, many potential drugs were explored. This led to several clinical trials for gentamicin and ataluren (PTC124) allowing stop codon read-through. An alternative approach is to induce the expression of an internally deleted, partially functional dystrophin protein through exon skipping. The vectors and the methods used in gene therapy have been continually improving in order to obtain greater encapsidation capacity and better transduction efficiency. The most promising experimental approaches using pharmaceutical and gene therapies are reviewed in this article. [ABSTRACT FROM AUTHOR]
- Published
- 2011
- Full Text
- View/download PDF
31. Prevention of Dystrophic Pathology in Severely Affected Dystrophin/Utrophin-deficient Mice by Morpholino-oligomer-mediated Exon-skipping.
- Author
-
Goyenvalle, Aurélie, Babbs, Arran, Powell, Dave, Kole, Ryszard, Fletcher, Sue, Wilton, Steve D., and Davies, Kay E.
- Subjects
- *
DUCHENNE muscular dystrophy , *NEUROMUSCULAR diseases , *DYSTROPHIN genes , *MEMBRANE proteins , *GENE expression , *EXONS (Genetics) - Abstract
Duchenne muscular dystrophy (DMD) is a severe neuromuscular disorder caused by mutations in the dystrophin gene that result in the absence of functional protein. Antisense-mediated exon-skipping is one of the most promising approaches for the treatment of DMD because of its capacity to correct the reading frame and restore dystrophin expression, which has been demonstrated in vitro and in vivo. In particular, peptide-conjugated phosphorodiamidate morpholino oligomers (PPMOs) have recently been shown to induce widespread high levels of dystrophin expression in the mdx mouse model. Here, we report the efficiency of the PPMO-mediated exon-skipping approach in the utrophin/dystrophin double-knockout mouse (dKO) mouse, which is a much more severe and progressive mouse model of DMD. Repeated intraperitoneal (i.p.) injections of a PPMO targeted to exon 23 of dystrophin pre-mRNA in dKO mice induce a near-normal level of dystrophin expression in all muscles examined, except for the cardiac muscle, resulting in a considerable improvement of their muscle function and dystrophic pathology. These findings suggest great potential for PPMOs in systemic treatment of the DMD phenotype. [ABSTRACT FROM AUTHOR]
- Published
- 2010
- Full Text
- View/download PDF
32. Enhanced Exon-skipping Induced by U7 snRNA Carrying a Splicing Silencer Sequence: Promising Tool for DMD Therapy.
- Author
-
Goyenvalle, Aurélie, Babbs, Arran, van Ommen, Gert-Jan B., Garcia, Luis, and Davies, Kay E.
- Subjects
- *
EXONS (Genetics) , *DUCHENNE muscular dystrophy , *MESSENGER RNA , *DYSTROPHIN genes , *GENE therapy , *OLIGONUCLEOTIDES , *DYSTROPHY - Abstract
Duchenne muscular dystrophy (DMD) is a fatal muscle wasting disorder caused by mutations in the dystrophin gene. In most cases, the open-reading frame is disrupted which results in the absence of functional protein. Antisense-mediated exon skipping is one of the most promising approaches for the treatment of DMD and has recently been shown to correct the reading frame and restore dystrophin expression in vitro and in vivo. Specific exon skipping can be achieved using synthetic oligonucleotides or viral vectors encoding modified small nuclear RNAs (snRNAs), by masking important splicing sites. In this study, we demonstrate that enhanced exon skipping can be induced by a U7 snRNA carrying binding sites for the heterogeneous ribonucleoprotein A1 (hnRNPA1). In DMD patient cells, bifunctional U7 snRNAs harboring silencer motifs induce complete skipping of exon 51, and thus restore dystrophin expression to near wild-type levels. Furthermore, we show the efficacy of these constructs in vivo in transgenic mice carrying the entire human DMD locus after intramuscular injection of adeno-associated virus (AAV) vectors encoding the bifunctional U7 snRNA. These new constructs are very promising for the optimization of therapeutic exon skipping for DMD, but also offer powerful and versatile tools to modulate pre-mRNA splicing in a wide range of applications.Molecular Therapy (2009) 17 7, 1234–1240. doi:10.1038/mt.2009.113 [ABSTRACT FROM AUTHOR]
- Published
- 2009
- Full Text
- View/download PDF
33. Generation and Characterization of Transgenic Mice with the Full-length Human DMD Gene.
- Author
-
't Hoen, Peter A. C., De Meijer, Emile J., Boer, Judith M., Vossen, Rolf H. A. M., Turk, Rolf, Maatman, Ronald G. H. J., Davies, Kay E., Van Ommen, Gert-Jan B., Van Deutekom, Judith C. T., and Den Dunnen, Johan T.
- Subjects
- *
DYSTROPHIN genes , *TRANSGENIC animals , *DNA , *BLASTOCYST , *CHROMOSOMES , *EMBRYONIC stem cells - Abstract
We report the generation of mice with an intact and functional copy of the 2.3-megabase human dystrophin gene (hDMD), the largest functional stretch of human DNA thus far integrated into a mouse chromosome. Yeast spheroplasts containing an artificial chromosome with the full-length hDMD gene were fused with mouse embryonic stem cells and were subsequently injected into mouse blastocysts to produce transgenic hDMD mice. Human-specific PCR, Southern blotting, and fluorescent in situ hybridization techniques demonstrated the intactness and stable chromosomal integration of the hDMD gene on mouse chromosome 5. Expression of the transgene was confirmed by RT-PCR and Western blotting. The tissue-specific expression pattern of the different DMD transcripts was maintained. However, the human Dp427p and Dp427m transcripts were expressed at 2-fold higher levels and human Dp427c and Dp260 transcripts were expressed at 2- and 4-fold lower levels than their endogenous counterparts. Ultimate functional proof of the hDMD transgene was obtained by crossing of hDMD mice with dystrophin-deficient mdx mice and dystrophin and utrophin-deficient mdx × Utrn-/- mice. The hDMD transgene rescued the lethal dystrophic phenotype of the mdx × Utrn-/- mice. All signs of muscular dystrophy disappeared in the rescued mice, as demonstrated by histological staining of muscle sections and gene expression profiling experiments. Currently, hDMD mice are extensively used for preclinical testing of sequence-specific therapeutics for the treatment of Duchenne muscular dystrophy. In addition, the hDMD mouse can be used to study the influence of the genomic context on deletion and recombination frequencies, genome stability, and gene expression regulation. [ABSTRACT FROM AUTHOR]
- Published
- 2008
- Full Text
- View/download PDF
34. Intermediate filament-like protein syncoilin in normal and myopathic striated muscle
- Author
-
McCullagh, Karl J.A., Edwards, Ben, Poon, Ellen, Lovering, Richard M., Paulin, Denise, and Davies, Kay E.
- Subjects
- *
MICE , *MUSCLE diseases , *MEMBRANE proteins , *DYSTROPHIN - Abstract
Abstract: The intermediate filament-like protein syncoilin is a member of the dystrophin protein complex, and links the complex to the cytoskeleton through binding α-dystrobrevin and desmin in muscle. Here, we identify further sites of syncoilin location in normal muscle: at the perinuclear space, myotendinous junction, and enrichment in the sarcolemma and sarcoplasm of oxidative muscle fibers in mice. To understand the importance of the dystrophin protein complex-syncoilin-cytoskeletal link and its implication to disease, we analyzed syncoilin in mice null for α-dystrobrevin (adbn−/−) and desmin (des−/−). Syncoilin was upregulated in dystrophic muscles of adbn−/− mice, without alteration in its subcellular location. In des−/− mice, syncoilin was severely reduced in skeletal muscle; lost from sarcomeric Z-lines and neuromuscular junctions, and redistributed from the sub-sarcolemmal cytoskeleton to the cytoplasm. The data show that absence of α-dystrobrevin or desmin leads to dynamic changes in syncoilin that may compensate for, or participate in, different muscle myopathies. [Copyright &y& Elsevier]
- Published
- 2007
- Full Text
- View/download PDF
35. Expression profiling in spinal muscular atrophy reveals an RNA binding protein deficit
- Author
-
Anderson, Kirstie N., Baban, Dilair, Oliver, Peter L., Potter, Allyson, and Davies, Kay E.
- Subjects
- *
SPINAL muscular atrophy , *CARRIER proteins , *PROTEINS , *CENTRAL nervous system - Abstract
Spinal muscular atrophy is a common neuromuscular disorder caused by deletions or mutations within the survival motor neuron gene. The reason for specific motor neuron loss within the disease is still unclear. Expression profiling has been carried out in two models of spinal muscular atrophy; the heterozygote mouse model and human primary muscle cultures from a spinal muscular atrophy patient. A group of RNA binding proteins are up-regulated in spinal muscular atrophy motor neurons. One such protein, BRUNOL3, is highly expressed within spinal cord and muscle and also at the same developmental stage as survival motor neuron. The differential expression of Brunol3 has been confirmed with real-time RT-PCR in spinal cord and muscle of three different models of spinal muscular atrophy. BRUNOL3 has been shown to co-localise with survival motor neuron in the nuclei of neuronal cells and to co-immunoprecipitate with Smn in mouse brain. This is the first time that a link has been established between RNA binding proteins and survival motor neuron within motor neurons. [Copyright &y& Elsevier]
- Published
- 2004
- Full Text
- View/download PDF
36. Isolation and culture of motor neurons from the newborn mouse spinal cord
- Author
-
Anderson, Kirstie N., Potter, Allyson C., Piccenna, Loretta G., Quah, Alvin K.J., Davies, Kay E., and Cheema, Surindar S.
- Subjects
- *
CELL separation , *MOTOR neurons , *MICROSCOPY , *CYTOLOGICAL techniques - Abstract
A protocol for the isolation and culture of motor neurons from postnatal day 1 mouse spinal cord is described. After 72 h in culture, phase contrast microscopy reveals healthy cells with motor neuronal morphology and extensive neuritic processes. These neurons express the 75-kDa low-affinity neurotrophin receptor (p75NTR) and choline acetyltransferase (ChAT), both proteins are specifically expressed by neonatal and embryonic motor neurons in vivo. This protocol can be adapted for various postnatal motor neuron assays. [Copyright &y& Elsevier]
- Published
- 2004
- Full Text
- View/download PDF
37. Syncoilin accumulation in two patients with desmin-related myopathy
- Author
-
Howman, Emily V., Sullivan, Nicky, Poon, Ellen P., Britton, Joanna E., Hilton-Jones, David, and Davies, Kay E.
- Subjects
- *
MUSCLE diseases , *MEMBRANE proteins , *DYSTROPHIN , *PROTEINS - Abstract
We have recently shown that syncoilin interacts with desmin in skeletal muscle and has a role in attaching and organising desmin filaments to the Z-lines. We have analysed patients with desmin accumulation and have found that syncoilin is both upregulated at the sarcolemma and aggregates with desmin indicating the presence of two distinct protein populations. Additional dystrophin-associated protein complex components also accumulate. The striking finding was that α-dystrobrevin-1 and neuronal nitric oxide synthase (nNOS) are almost completely lost from the membrane of these patients indicating that the myopathy may result from both the abnormal accumulation of proteins and an increase in ischaemic injury due to the loss of nNOS. We speculate that the loss of α-dystrobrevin from the membrane, and subsequent loss of nNOS, is due to the α-dystrobrevin–syncoilin–desmin interaction. [Copyright &y& Elsevier]
- Published
- 2003
- Full Text
- View/download PDF
38. Discovery and mechanism of action studies of 4,6-diphenylpyrimidine-2-carbohydrazides as utrophin modulators for the treatment of Duchenne muscular dystrophy.
- Author
-
Vuorinen, Aini, Wilkinson, Isabel V.L., Chatzopoulou, Maria, Edwards, Ben, Squire, Sarah E., Fairclough, Rebecca J., Bazan, Noelia Araujo, Milner, Josh A., Conole, Daniel, Donald, James R., Shah, Nandini, Willis, Nicky J., Martínez, R. Fernando, Wilson, Francis X., Wynne, Graham M., Davies, Stephen G., Davies, Kay E., and Russell, Angela J.
- Subjects
- *
DUCHENNE muscular dystrophy , *PROTEIN expression , *PROTEOMICS , *CYTOSKELETAL proteins , *ANALYTICAL chemistry , *PHENOTYPES - Abstract
Duchenne muscular dystrophy is a fatal disease with no cure, caused by lack of the cytoskeletal protein dystrophin. Upregulation of utrophin, a dystrophin paralogue, offers a potential therapy independent of mutation type. The failure of first-in-class utrophin modulator ezutromid/SMT C1100 in Phase II clinical trials necessitates development of compounds with better efficacy, physicochemical and ADME properties and/or complementary mechanisms. We have discovered and performed a preliminary optimisation of a novel class of utrophin modulators using an improved phenotypic screen, where reporter expression is derived from the full genomic context of the utrophin promoter. We further demonstrate through target deconvolution studies, including expression analysis and chemical proteomics, that this compound series operates via a novel mechanism of action, distinct from that of ezutromid. [Display omitted] • Discovery of a new class of utrophin modulators for Duchenne muscular dystrophy. • Development of an improved phenotypic screen derived from a reporter knock-in mouse. • Target deconvoluted using photoaffinity chemical proteomics and expression analysis. • Mechanism of action is distinct from previously reported utrophin modulators. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
39. Muscling in on gene therapy
- Author
-
Davies, Kay E.
- Published
- 1996
- Full Text
- View/download PDF
40. Translating the Genomics Revolution: The Need for an International Gene Therapy Consortium for Monogenic Diseases.
- Author
-
Tremblay, Jacques P, Xiao, Xiao, Aartsma-Rus, Annemieke, Barbas, Carlos, Blau, Helen M, Bogdanove, Adam J, Boycott, Kym, Braun, Serge, Breakefield, Xandra O, Bueren, Juan A, Buschmann, Michael, Byrne, Barry J, Calos, Michele, Cathomen, Toni, Chamberlain, Jeffrey, Chuah, Marinee, Cornetta, Kenneth, Davies, Kay E, Dickson, J George, and Duchateau, Philippe
- Subjects
- *
LETTERS to the editor , *GENE therapy , *GENETIC disorders - Abstract
A letter to the editor which stresses the need for an international gene therapy consortium for monogenic diseases is presented.
- Published
- 2013
- Full Text
- View/download PDF
41. Alterations of neuromuscular junctions in Duchenne muscular dystrophy.
- Author
-
Lovering, Richard M., Iyer, Shama R., Edwards, Benjamin, and Davies, Kay E.
- Subjects
- *
DUCHENNE muscular dystrophy , *MYONEURAL junction , *NEUROMUSCULAR transmission , *MUSCULAR dystrophy , *MUSCLE weakness , *FACIOSCAPULOHUMERAL muscular dystrophy - Abstract
• Duchenne muscular dystrophy (DMD), the most common form of muscular dystrophy, is caused by the lack of dystrophin, a protein encoded by the DMD gene. • The neuromuscular junction (NMJ) is not a fixed, permanent structure, but instead shows plasticity in response to injury, exercise, and aging. • NMJs in the mdx mouse model of DMD show aberrant changes in both pre- and post-synaptic NMJ structure, which could influence neuromuscular transmission. • The hypothesis that the NMJ contributes to functional deficits in DMD represents a paradigm shift from more prevalent myo-centric perspectives. The focus of this review is on Duchenne muscular dystrophy (DMD), which is caused by the absence of the protein dystrophin and is characterized as a neuromuscular disease in which muscle weakness, increased susceptibility to muscle injury, and inadequate repair appear to underlie the pathology. Considerable attention has been dedicated to studying muscle fiber damage, but data show that both human patients and animal models for DMD present with fragmented neuromuscular junction (NMJ) morphology. In addition to pre- and post-synaptic abnormalities, studies indicate increased susceptibility of the NMJ to contraction-induced injury, with corresponding functional changes in neuromuscular transmission and nerve-evoked electromyographic activity. Such findings suggest that alterations in the NMJ of dystrophic muscle may play a role in muscle weakness via impairment of neuromuscular transmission. Further work is needed to fully understand the role of the NMJ in the weakness, susceptibility to injury, and progressive wasting associated with DMD. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
42. Synthesis of SMT022357 enantiomers and in vivo evaluation in a Duchenne muscular dystrophy mouse model.
- Author
-
Babbs, Arran, Berg, Adam, Chatzopoulou, Maria, Davies, Kay E., Davies, Stephen G., Edwards, Benjamin, Elsey, David J., Emer, Enrico, Figuccia, Aude L.A., Fletcher, Ai M., Guiraud, Simon, Harriman, Shawn, Moir, Lee, Robinson, Neil, Rowley, Jessica A., Russell, Angela J., Squire, Sarah E., Thomson, James E., Tinsley, Jonathon M., and Wilson, Francis X.
- Subjects
- *
DUCHENNE muscular dystrophy , *ENANTIOMERS , *RACEMIC mixtures , *ASYMMETRIC synthesis , *MICE - Abstract
Following on from ezutromid, the first-in-class benzoxazole utrophin modulator that progressed to Phase 2 clinical trials for the treatment of Duchenne muscular dystrophy, a new chemotype was designed to optimise its physicochemical and ADME profile. Herein we report the synthesis of SMT022357, a second generation utrophin modulator preclinical candidate, and an asymmetric synthesis of its constituent enantiomers. The pharmacological properties of both enantiomers were evaluated in vitro and in vivo. No significant difference in the activity or efficacy was observed between the two enantiomers; activity was found to be comparable to the racemic mixture. Image 1 • A stereoselective synthesis was devised to access enantiomers of phosphinate SMT022357. • SMT022357 enantiomers have an improved ADME profile compared to ezutromid. • SMT022357 enantiomers show comparable activity and improve the dystrophic phenotype in mdx mice. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.