Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 158
Filter
1.
Am J Med Genet A ; 194(6): e63514, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38329159

ABSTRACT

Genetics has become a critical component of medicine over the past five to six decades. Alongside genetics, a relatively new discipline, dysmorphology, has also begun to play an important role in providing critically important diagnoses to individuals and families. Both have become indispensable to unraveling rare diseases. Almost every medical specialty relies on individuals experienced in these specialties to provide diagnoses for patients who present themselves to other doctors. Additionally, both specialties have become reliant on molecular geneticists to identify genes associated with human disorders. Many of the medical geneticists, dysmorphologists, and molecular geneticists traveled a circuitous route before arriving at the position they occupied. The purpose of collecting the memoirs contained in this article was to convey to the reader that many of the individuals who contributed to the advancement of genetics and dysmorphology since the late 1960s/early 1970s traveled along a journey based on many chances taken, replying to the necessities they faced along the way before finding full enjoyment in the practice of medical and human genetics or dysmorphology. Additionally, and of equal importance, all exhibited an ability to evolve with their field of expertise as human genetics became human genomics with the development of novel technologies.


Subject(s)
Genetics, Medical , Humans , History, 20th Century , History, 21st Century , Human Genetics
2.
Nucleic Acids Res ; 50(10): 5443-5466, 2022 06 10.
Article in English | MEDLINE | ID: mdl-35061895

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.


Subject(s)
Muscular Dystrophy, Duchenne , Oligonucleotides, Antisense/chemistry , Phosphorothioate Oligonucleotides/chemistry , Animals , Exons , Mice , Muscle, Skeletal , Muscular Dystrophy, Duchenne/drug therapy , Muscular Dystrophy, Duchenne/therapy , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/pharmacology , Phosphorothioate Oligonucleotides/pharmacology , RNA Splicing/drug effects
3.
Annu Rev Genomics Hum Genet ; 21: 1-13, 2020 08 31.
Article in English | MEDLINE | ID: mdl-32119789

ABSTRACT

I was honored to be asked by the Editorial Committee of the Annual Review of Genomics and Genetics to write an autobiographical account of my life in science and in genetics in particular. The field has moved from mapping Mendelian disorders 40 years ago to the delivery of effective therapies for some monogenic disorders today. My 40-year journey from diagnosis to therapy for Duchenne muscular dystrophy has depended on collaborations among basic scientists, clinicians, medical charities, genetic counselors, biotech companies, and affected families. The future of human genetics looks even more exciting, with techniques such as single-cell sequencing and somatic cell CRISPR editing opening up opportunities for precision medicine and accelerating progress.


Subject(s)
Disease Models, Animal , Dystrophin/genetics , Genetic Therapy , Muscular Dystrophy, Duchenne/diagnosis , Muscular Dystrophy, Duchenne/therapy , Mutation , Animals , Gene Editing , History, 20th Century , History, 21st Century , Humans , Periodicals as Topic
4.
Cell ; 133(4): 572-4, 2008 May 16.
Article in English | MEDLINE | ID: mdl-18485864

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, Zhang et al. (2008) challenge prior assumptions about the housekeeping function of SMN and demonstrate that loss of SMN leads to highly tissue-specific effects on splicing.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Motor Neurons/metabolism , Nerve Tissue Proteins/metabolism , RNA Splicing , RNA-Binding Proteins/metabolism , Animals , Humans , Mice , Muscular Atrophy, Spinal/metabolism , Ribonucleoproteins, Small Nuclear/metabolism , SMN Complex Proteins
5.
Bioorg Med Chem ; 69: 116812, 2022 09 01.
Article in English | MEDLINE | ID: mdl-35772287

ABSTRACT

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.


Subject(s)
Muscular Dystrophy, Duchenne , Animals , Hydrazines/pharmacology , Hydrazines/therapeutic use , Mice , Muscle, Skeletal/metabolism , Muscular Dystrophy, Duchenne/drug therapy , Muscular Dystrophy, Duchenne/metabolism , Structure-Activity Relationship , Up-Regulation , Utrophin/genetics , Utrophin/metabolism , Utrophin/therapeutic use
6.
Hum Mutat ; 42(1): 3-7, 2021 01.
Article in English | MEDLINE | ID: mdl-33252176

ABSTRACT

Documenting variation in our genomes is important for research and clinical care. Accuracy in the description of DNA variants is therefore essential. To address this issue, the Human Variome Project convened a committee to evaluate the feasibility of requiring authors to verify that all variants submitted for publication complied with a widely accepted standard for description. After a pilot study of two journals, the committee agreed that requiring authors to verify that variants complied with Human Genome Variation Society nomenclature is a reasonable step toward standardizing the worldwide inventory of human variation.


Subject(s)
DNA , Genome, Human , Periodicals as Topic , Terminology as Topic , DNA/genetics , Genetic Variation , Human Genome Project , Humans , Periodicals as Topic/standards , Pilot Projects , Publications/standards
7.
Hum Mol Genet ; 28(13): 2189-2200, 2019 07 01.
Article in English | MEDLINE | ID: mdl-30990876

ABSTRACT

Duchenne muscular dystrophy (DMD) is a lethal neuromuscular disorder caused by loss of dystrophin. Several therapeutic modalities are currently in clinical trials but none will achieve maximum functional rescue and full disease correction. Therefore, we explored the potential of combining the benefits of dystrophin with increases of utrophin, an autosomal paralogue of dystrophin. Utrophin and dystrophin can be co-expressed and co-localized at the same muscle membrane. Wild-type (wt) levels of dystrophin are not significantly affected by a moderate increase of utrophin whereas higher levels of utrophin reduce wt dystrophin, suggesting a finite number of actin binding sites at the sarcolemma. Thus, utrophin upregulation strategies may be applied to the more mildly affected Becker patients with lower dystrophin levels. Whereas increased dystrophin in wt animals does not offer functional improvement, overexpression of utrophin in wt mice results in a significant supra-functional benefit over wt. These findings highlight an additive benefit of the combined therapy and potential new unique roles of utrophin. Finally, we show a 30% restoration of wt dystrophin levels, using exon-skipping, together with increased utrophin levels restores dystrophic muscle function to wt levels offering greater therapeutic benefit than either single approach alone. Thus, this combination therapy results in additive functional benefit and paves the way for potential future combinations of dystrophin- and utrophin-based strategies.


Subject(s)
Dystrophin/genetics , Muscular Dystrophy, Duchenne/therapy , Utrophin/genetics , Animals , Dystrophin/metabolism , Exons , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Morpholinos/chemical synthesis , Morpholinos/therapeutic use , Muscle, Skeletal/metabolism , Muscular Dystrophy, Duchenne/genetics , Myofibrils/metabolism , Sarcolemma/metabolism , Up-Regulation , Utrophin/metabolism
8.
Hum Mol Genet ; 28(21): 3584-3599, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31642482

ABSTRACT

A common pathological hallmark of amyotrophic lateral sclerosis (ALS) and the related neurodegenerative disorder frontotemporal dementia, is the cellular mislocalization of transactive response DNA-binding protein 43 kDa (TDP-43). Additionally, multiple mutations in the TARDBP gene (encoding TDP-43) are associated with familial forms of ALS. While the exact role for TDP-43 in the onset and progression of ALS remains unclear, the identification of factors that can prevent aberrant TDP-43 localization and function could be clinically beneficial. Previously, we discovered that the oxidation resistance 1 (Oxr1) protein could alleviate cellular mislocalization phenotypes associated with TDP-43 mutations, and that over-expression of Oxr1 was able to delay neuromuscular abnormalities in the hSOD1G93A ALS mouse model. Here, to determine whether Oxr1 can protect against TDP-43-associated phenotypes in vitro and in vivo, we used the same genetic approach in a newly described transgenic mouse expressing the human TDP-43 locus harbouring an ALS disease mutation (TDP-43M337V). We show in primary motor neurons from TDP-43M337V mice that genetically-driven Oxr1 over-expression significantly alleviates cytoplasmic mislocalization of mutant TDP-43. We also further quantified newly-identified, late-onset neuromuscular phenotypes of this mutant line, and demonstrate that neuronal Oxr1 over-expression causes a significant reduction in muscle denervation and neuromuscular junction degeneration in homozygous mutants in parallel with improved motor function and a reduction in neuroinflammation. Together these data support the application of Oxr1 as a viable and safe modifier of TDP-43-associated ALS phenotypes.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , DNA-Binding Proteins/metabolism , Mitochondrial Proteins/metabolism , Motor Neurons/metabolism , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Amyotrophic Lateral Sclerosis/prevention & control , Animals , Cytoplasm/metabolism , DNA-Binding Proteins/genetics , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mitochondrial Proteins/genetics , Muscle Denervation , Muscles/innervation , Mutation, Missense , Neuromuscular Junction/metabolism , Protein Transport
9.
Hum Mol Genet ; 28(2): 307-319, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30304405

ABSTRACT

Duchenne muscular dystrophy (DMD) is a lethal, X-linked muscle-wasting disease caused by lack of the cytoskeletal protein dystrophin. Constitutive utrophin expression, a structural and functional paralogue of dystrophin, can successfully prevent the dystrophic pathology in the dystrophin-deficient mdx mouse model. In dystrophic muscles, utrophin is increased as part of the repair process and localized at the sarcolemma of regenerating myofibers. The presence of developmental myosin such as embryonic myosin (MyHC-emb) and neonatal represents a useful marker of muscle regeneration and a meaningful indicator of muscle damage, which correlates with the clinical severity of milder Becker muscular dystrophy and DMD patients. In the present study, we demonstrate that MyHC-emb is a robust marker of regeneration at different ages and in different skeletal muscles. We also evaluate the correlation between utrophin, dystrophin and MyHC-emb in wild-type (wt) and regenerating dystrophic muscles. Restoration of dystrophin significantly reduced MyHC-emb levels. Similarly, overexpression of utrophin in the transgenic mdx-Fiona mice reduced the number of MyHC-emb positive fibers to wt level, prevented the regenerative process and rescued the muscle function. In contrast, the absence of utrophin in the dystrophin-deficient double-knockout mice resulted in a higher MyHC-emb content and in a more severe dystrophic pathophysiology than in mdx mice. These data illustrate the importance of monitoring utrophin and MyHC-emb levels in the preclinical evaluation of therapies and provide translational support for the use of developmental myosin as a disease biomarker in DMD clinical trials.


Subject(s)
Dystrophin/metabolism , Muscle, Skeletal/metabolism , Muscular Dystrophy, Duchenne/metabolism , Myosins/metabolism , Regeneration , Utrophin/metabolism , Animals , Biomarkers/metabolism , Clinical Trials as Topic , Disease Models, Animal , Embryo, Mammalian/metabolism , Male , Mice, Inbred C57BL , Mice, Inbred mdx , Mice, Transgenic , Muscle, Skeletal/embryology , Muscle, Skeletal/physiology , Muscular Dystrophy, Animal , Muscular Dystrophy, Duchenne/drug therapy , Muscular Dystrophy, Duchenne/embryology , Muscular Dystrophy, Duchenne/pathology , Sarcolemma/metabolism
10.
Biochem Soc Trans ; 48(3): 813-821, 2020 06 30.
Article in English | MEDLINE | ID: mdl-32597486

ABSTRACT

Genetic approaches for the diagnosis and treatment of inherited muscle diseases have advanced rapidly in recent years. Many of the advances have occurred in the treatment of Duchenne muscular dystrophy (DMD), a muscle wasting disease where affected boys are typically wheelchair bound by age 12 years and generally die in their twenties from respiratory failure or cardiomyopathy. Dystrophin is a 421 kD protein which links F-actin to the extracellular matrix via the dystrophin-associated protein complex (DAPC) at the muscle membrane. In the absence of dystrophin, the DAPC is lost, making the muscle membrane more susceptible to contraction-induced injury. The identification of the gene causing DMD in 1986 resulted in improved diagnosis of the disease and the identification of hotspots for mutation. There is currently no effective treatment. However, there are several promising genetic therapeutic approaches at the preclinical stage or in clinical trials including read-through of stop codons, exon skipping, delivery of dystrophin minigenes and the modulation of expression of the dystrophin related protein, utrophin. In spite of significant progress, the problem of targeting all muscles, including diaphragm and heart at sufficiently high levels, remains a challenge. Any therapy also needs to consider the immune response and some treatments are mutation specific and therefore limited to a subgroup of patients. This short review provides a summary of the current status of DMD therapy with a particular focus on those genetic strategies that have been taken to the clinic.


Subject(s)
Muscular Dystrophy, Duchenne/diagnosis , Muscular Dystrophy, Duchenne/therapy , CRISPR-Cas Systems , Codon, Terminator , Dystrophin/genetics , Exons , Genetic Therapy/methods , Humans , Male , Muscular Dystrophy, Duchenne/genetics
11.
Tetrahedron ; 76(2): 130819, 2020 Jan 10.
Article in English | MEDLINE | ID: mdl-32713969

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.

12.
Angew Chem Int Ed Engl ; 59(6): 2420-2428, 2020 02 03.
Article in English | MEDLINE | ID: mdl-31755636

ABSTRACT

Duchenne muscular dystrophy (DMD) is a fatal muscle-wasting disease arising from mutations in the dystrophin gene. Upregulation of utrophin to compensate for the missing dystrophin offers a potential therapy independent of patient genotype. The first-in-class utrophin modulator ezutromid/SMT C1100 was developed from a phenotypic screen through to a Phase 2 clinical trial. Promising efficacy and evidence of target engagement was observed in DMD patients after 24 weeks of treatment, however trial endpoints were not met after 48 weeks. The objective of this study was to understand the mechanism of action of ezutromid which could explain the lack of sustained efficacy and help development of new generations of utrophin modulators. Using chemical proteomics and phenotypic profiling we show that the aryl hydrocarbon receptor (AhR) is a target of ezutromid. Several lines of evidence demonstrate that ezutromid binds AhR with an apparent KD of 50 nm and behaves as an AhR antagonist. Furthermore, other reported AhR antagonists also upregulate utrophin, showing that this pathway, which is currently being explored in other clinical applications including oncology and rheumatoid arthritis, could also be exploited in future DMD therapies.


Subject(s)
Benzoxazoles/chemistry , Naphthalenes/chemistry , Proteomics/methods , Receptors, Aryl Hydrocarbon/metabolism , Utrophin/metabolism , Animals , Benzoxazoles/metabolism , Benzoxazoles/pharmacology , Benzoxazoles/therapeutic use , Cycloaddition Reaction , Drug Design , Humans , Kinetics , Mice , Molecular Probes/chemistry , Muscular Dystrophy, Duchenne/drug therapy , Myoblasts/cytology , Myoblasts/metabolism , Naphthalenes/metabolism , Naphthalenes/pharmacology , Naphthalenes/therapeutic use , Protein Binding , Receptors, Aryl Hydrocarbon/antagonists & inhibitors , Receptors, Aryl Hydrocarbon/genetics , Up-Regulation/drug effects , Utrophin/agonists , Utrophin/genetics
13.
Nat Rev Genet ; 14(6): 373-8, 2013 06.
Article in English | MEDLINE | ID: mdl-23609411

ABSTRACT

Duchenne muscular dystrophy (DMD) is a devastating progressive disease for which there is currently no effective treatment except palliative therapy. There are several promising genetic approaches, including viral delivery of the missing dystrophin gene, read-through of translation stop codons, exon skipping to restore the reading frame and increased expression of the compensatory utrophin gene. The lessons learned from these approaches will be applicable to many other disorders.


Subject(s)
Genetic Therapy , Muscular Dystrophy, Duchenne/therapy , Animals , Clinical Trials as Topic , Dependovirus/genetics , Dystrophin/chemistry , Dystrophin/genetics , Dystrophin/metabolism , Gene Expression , Genetic Vectors , Humans , Muscular Dystrophy, Duchenne/genetics , Mutation , Protein Structure, Tertiary , Utrophin/genetics , Utrophin/metabolism
14.
Hum Mol Genet ; 30(20): R155, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34608484
15.
Annu Rev Genomics Hum Genet ; 16: 281-308, 2015.
Article in English | MEDLINE | ID: mdl-26048046

ABSTRACT

Current molecular genomic approaches to human genetic disorders have led to an explosion in the identification of the genes and their encoded proteins responsible for these disorders. The identification of the gene altered by mutations in Duchenne and Becker muscular dystrophy was one of the earliest examples of this paradigm. The nearly 30 years of research partly outlined here exemplifies the road that similar current gene discovery protocols will be expected to travel, albeit much more rapidly owing to improved diagnosis of genetic disorders and an understanding of the spectrum of mutations thought to cause them. The identification of the protein dystrophin has led to a new understanding of the muscle cell membrane and the proteins involved in membrane stability, as well as new candidate genes for additional forms of muscular dystrophy. Animal models identified with naturally occurring mutations and developed by genetic manipulation have furthered the understanding of disease progression and underlying pathology. The biochemistry and molecular analysis of patient samples have led to the different dystrophin-dependent and -independent therapies that are currently close to or in human clinical trials. The lessons learned from decades of research on dystrophin have benefited the field of human genetics.


Subject(s)
Dystrophin/metabolism , Muscular Dystrophies/physiopathology , Muscular Dystrophies/therapy , Animals , Clinical Trials as Topic , Disease Models, Animal , Dystrophin/genetics , Genetic Therapy/methods , Humans , Molecular Targeted Therapy/methods , Mutation , Phosphoric Diester Hydrolases/metabolism , Steroids/therapeutic use , Utrophin/genetics , Utrophin/metabolism
16.
Nature ; 484(7394): 394-8, 2012 Apr 04.
Article in English | MEDLINE | ID: mdl-22495301

ABSTRACT

Duchenne muscular dystrophy (DMD) is a severe and progressive muscle wasting disorder caused by mutations in the dystrophin gene that result in the absence of the membrane-stabilizing protein dystrophin. Dystrophin-deficient muscle fibres are fragile and susceptible to an influx of Ca(2+), which activates inflammatory and muscle degenerative pathways. At present there is no cure for DMD, and existing therapies are ineffective. Here we show that increasing the expression of intramuscular heat shock protein 72 (Hsp72) preserves muscle strength and ameliorates the dystrophic pathology in two mouse models of muscular dystrophy. Treatment with BGP-15 (a pharmacological inducer of Hsp72 currently in clinical trials for diabetes) improved muscle architecture, strength and contractile function in severely affected diaphragm muscles in mdx dystrophic mice. In dko mice, a phenocopy of DMD that results in severe spinal curvature (kyphosis), muscle weakness and premature death, BGP-15 decreased kyphosis, improved the dystrophic pathophysiology in limb and diaphragm muscles and extended lifespan. We found that the sarcoplasmic/endoplasmic reticulum Ca(2+)-ATPase (SERCA, the main protein responsible for the removal of intracellular Ca(2+)) is dysfunctional in severely affected muscles of mdx and dko mice, and that Hsp72 interacts with SERCA to preserve its function under conditions of stress, ultimately contributing to the decreased muscle degeneration seen with Hsp72 upregulation. Treatment with BGP-15 similarly increased SERCA activity in dystrophic skeletal muscles. Our results provide evidence that increasing the expression of Hsp72 in muscle (through the administration of BGP-15) has significant therapeutic potential for DMD and related conditions, either as a self-contained therapy or as an adjuvant with other potential treatments, including gene, cell and pharmacological therapies.


Subject(s)
Disease Progression , HSP72 Heat-Shock Proteins/metabolism , Muscle, Skeletal/physiology , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/physiopathology , Animals , Calcium-Transporting ATPases/metabolism , Diaphragm/drug effects , Diaphragm/physiology , Disease Models, Animal , Female , Gene Expression Regulation/drug effects , HSP72 Heat-Shock Proteins/biosynthesis , HSP72 Heat-Shock Proteins/genetics , Kyphosis/drug therapy , Longevity/drug effects , Male , Mice , Mice, Inbred mdx , Mice, Transgenic , Muscle, Skeletal/drug effects , Muscle, Skeletal/physiopathology , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/pathology , Oximes/pharmacology , Piperidines/pharmacology , Rats
17.
J Biol Chem ; 291(6): 2751-63, 2016 Feb 05.
Article in English | MEDLINE | ID: mdl-26668325

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.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Carrier Proteins/metabolism , Evolution, Molecular , Mitochondrial Proteins/metabolism , Neuroprotective Agents/metabolism , Nuclear Proteins/metabolism , Oxidative Stress , Amino Acid Motifs , Amyotrophic Lateral Sclerosis/genetics , Animals , Carrier Proteins/genetics , Cell Line , Disease Models, Animal , GTPase-Activating Proteins , INDEL Mutation , Mice , Mitochondrial Proteins/genetics , Nuclear Proteins/genetics , Protein Structure, Tertiary
18.
Hum Mol Genet ; 24(12): 3529-44, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-25792726

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease characterized by the loss of motor neuron-like cells. Mutations in the RNA- and DNA-binding proteins, fused in sarcoma (FUS) and transactive response DNA-binding protein 43 kDa (TDP-43), are responsible for 5-10% of familial and 1% of sporadic ALS cases. Importantly, aggregation of misfolded FUS or TDP-43 is also characteristic of several neurodegenerative disorders in addition to ALS, including frontotemporal lobar degeneration. Moreover, splicing deregulation of FUS and TDP-43 target genes as well as mitochondrial abnormalities are associated with disease-causing FUS and TDP-43 mutants. While progress has been made to understand the functions of these proteins, the exact mechanisms by which FUS and TDP-43 cause ALS remain unknown. Recently, we discovered that, in addition to being up-regulated in spinal cords of ALS patients, the novel protein oxidative resistance 1 (Oxr1) protects neurons from oxidative stress-induced apoptosis. To further understand the function of Oxr1, we present here the first interaction study of the protein. We show that Oxr1 binds to Fus and Tdp-43 and that certain ALS-associated mutations in Fus and Tdp-43 affect their Oxr1-binding properties. We further demonstrate that increasing Oxr1 levels in cells expressing specific Fus and Tdp-43 mutants improves the three main cellular features associated with ALS: cytoplasmic mis-localization and aggregation, splicing changes of a mitochondrial gene and mitochondrial defects. Taken together, these findings suggest that OXR1 may have therapeutic benefits for the treatment of ALS and related neurodegenerative disorders with TDP-43 pathology.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , DNA-Binding Proteins/genetics , Mutation , Proteins/genetics , Proteins/metabolism , RNA-Binding Protein FUS/genetics , Animals , Arginine/metabolism , Autophagy/genetics , Cytoplasm/metabolism , Humans , Methylation , Mice , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Oxidative Stress , Proteasome Endopeptidase Complex/metabolism , Protein Aggregation, Pathological , Protein Binding , Protein Interaction Domains and Motifs , Protein Isoforms , Proteins/chemistry , Proteolysis , RNA Splicing , Transcription, Genetic
19.
Hum Mol Genet ; 24(14): 4114-25, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-25908616

ABSTRACT

The Moonwalker (Mwk) mouse is a model of dominantly inherited cerebellar ataxia caused by a gain-of-function mutation in the transient receptor potential (TRP) channel TRPC3. Here, we report impairments in dendritic growth and synapse formation early on during Purkinje cell development in the Mwk cerebellum that are accompanied by alterations in calcium signaling. To elucidate the molecular effector pathways that regulate Purkinje cell dendritic arborization downstream of mutant TRPC3, we employed transcriptomic analysis of developing Purkinje cells isolated by laser-capture microdissection. We identified significant gene and protein expression changes in molecules involved in lipid metabolism. Consistently, lipid homeostasis in the Mwk cerebellum was found to be disturbed, and treatment of organotypic cerebellar slices with ceramide significantly improved dendritic outgrowth of Mwk Purkinje cells. These findings provide the first mechanistic insights into the TRPC3-dependent mechanisms, by which activated calcium signaling is coupled to lipid metabolism and the regulation of Purkinje cell development in the Mwk cerebellum.


Subject(s)
Calcium Signaling , Cerebellar Ataxia/genetics , Cerebellum/physiology , Lipid Metabolism , TRPC Cation Channels/metabolism , Animals , Cerebellar Ataxia/pathology , Cerebellum/metabolism , Dendrites/metabolism , Gene Expression Regulation , Mice , Purkinje Cells/metabolism , TRPC Cation Channels/genetics , Transcriptome
20.
Hum Mol Genet ; 24(15): 4212-24, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-25935002

ABSTRACT

Duchenne muscular dystrophy (DMD) is a lethal, X-linked muscle-wasting disease caused by lack of the cytoskeletal protein dystrophin. There is currently no cure for DMD although various promising approaches are progressing through human clinical trials. By pharmacologically modulating the expression of the dystrophin-related protein utrophin, we have previously demonstrated in dystrophin-deficient mdx studies, daily SMT C1100 treatment significantly reduced muscle degeneration leading to improved muscle function. This manuscript describes the significant disease modifying benefits associated with daily dosing of SMT022357, a second-generation compound in this drug series with improved physicochemical properties and a more robust metabolism profile. These studies in the mdx mouse demonstrate that oral administration of SMT022357 leads to increased utrophin expression in skeletal, respiratory and cardiac muscles. Significantly, utrophin expression is localized along the length of the muscle fibre, not just at the synapse, and is fibre-type independent, suggesting that drug treatment is modulating utrophin transcription in extra-synaptic myonuclei. This results in improved sarcolemmal stability and prevents dystrophic pathology through a significant reduction of regeneration, necrosis and fibrosis. All these improvements combine to protect the mdx muscle from contraction induced damage and enhance physiological function. This detailed evaluation of the SMT C1100 drug series strongly endorses the therapeutic potential of utrophin modulation as a disease modifying therapeutic strategy for all DMD patients irrespective of their dystrophin mutation.


Subject(s)
Dystrophin/biosynthesis , Muscle Fibers, Skeletal/drug effects , Muscular Dystrophy, Duchenne/drug therapy , Utrophin/biosynthesis , Animals , Dystrophin/genetics , Gene Expression Regulation/drug effects , Humans , Mice , Mice, Inbred mdx , Muscle Contraction/drug effects , Muscle Contraction/genetics , Muscle Fibers, Skeletal/pathology , Muscles/drug effects , Muscles/pathology , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/pathology , Sarcolemma/drug effects , Sarcolemma/genetics , Utrophin/genetics
SELECTION OF CITATIONS
SEARCH DETAIL