Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
RNA ; 29(4): 489-497, 2023 04.
Article in English | MEDLINE | ID: mdl-36693761

ABSTRACT

Disruptions in RNA processing play critical roles in the pathogenesis of neurological diseases. In this Perspective, we discuss recent progress in the development of RNA-targeting therapeutic modalities. We focus on progress, limitations, and opportunities in a new generation of therapies engineered from RNA binding proteins and other endogenous RNA regulatory macromolecules to treat human neurological disorders.


Subject(s)
Nervous System Diseases , RNA , Humans , RNA/genetics , Nervous System Diseases/drug therapy , Nervous System Diseases/genetics , RNA Processing, Post-Transcriptional
2.
Nat Neurosci ; 26(1): 27-38, 2023 01.
Article in English | MEDLINE | ID: mdl-36510111

ABSTRACT

Huntington's disease (HD) is a fatal, dominantly inherited neurodegenerative disorder caused by CAG trinucleotide expansion in exon 1 of the huntingtin (HTT) gene. Since the reduction of pathogenic mutant HTT messenger RNA is therapeutic, we developed a mutant allele-sensitive CAGEX RNA-targeting CRISPR-Cas13d system (Cas13d-CAGEX) that eliminates toxic CAGEX RNA in fibroblasts derived from patients with HD and induced pluripotent stem cell-derived neurons. We show that intrastriatal delivery of Cas13d-CAGEX via an adeno-associated viral vector selectively reduces mutant HTT mRNA and protein levels in the striatum of heterozygous zQ175 mice, a model of HD. This also led to improved motor coordination, attenuated striatal atrophy and reduction of mutant HTT protein aggregates. These phenotypic improvements lasted for at least eight months without adverse effects and with minimal off-target transcriptomic effects. Taken together, we demonstrate proof of principle of an RNA-targeting CRISPR-Cas13d system as a therapeutic approach for HD, a strategy with implications for the treatment of other dominantly inherited disorders.


Subject(s)
Huntington Disease , Mice , Animals , Huntington Disease/genetics , Huntington Disease/therapy , Huntington Disease/metabolism , RNA , Clustered Regularly Interspaced Short Palindromic Repeats , Corpus Striatum/metabolism , RNA, Messenger/metabolism , Phenotype , Huntingtin Protein/genetics , Huntingtin Protein/metabolism , Disease Models, Animal
3.
Sci Transl Med ; 14(651): eabn2375, 2022 06 29.
Article in English | MEDLINE | ID: mdl-35767654

ABSTRACT

Myotonic dystrophy type 1 (DM1) is a multisystem, autosomal-dominant inherited disorder caused by CTG microsatellite repeat expansions (MREs) in the 3' untranslated region of the dystrophia myotonica-protein kinase (DMPK) gene. Despite its prominence as the most common adult-onset muscular dystrophy, patients with congenital to juvenile-onset forms of DM1 can present with debilitating neurocognitive symptoms along the autism spectrum, characteristic of possible in utero cortical defects. However, the molecular mechanism by which CTG MREs lead to these developmental central nervous system (CNS) manifestations is unknown. Here, we showed that CUG foci found early in the maturation of three-dimensional (3D) cortical organoids from DM1 patient-derived induced pluripotent stem cells (iPSCs) cause hyperphosphorylation of CUGBP Elav-like family member 2 (CELF2) protein. Integrative single-cell RNA sequencing and enhanced cross-linking and immunoprecipitation (eCLIP) analysis revealed that reduced CELF2 protein-RNA substrate interactions results in misregulation of genes critical for excitatory synaptic signaling in glutamatergic neurons, including key components of the methyl-CpG binding protein 2 (MECP2) pathway. Comparisons to MECP2(y/-) cortical organoids revealed convergent molecular and cellular defects such as glutamate toxicity and neuronal loss. Our findings provide evidence suggesting that early-onset DM1 might involve neurodevelopmental disorder-associated pathways and identify N-methyl-d-aspartic acid (NMDA) antagonists as potential treatment avenues for neuronal defects in DM1.


Subject(s)
Methyl-CpG-Binding Protein 2 , Myotonic Dystrophy , Adult , CELF Proteins/genetics , CELF Proteins/metabolism , Humans , Methyl-CpG-Binding Protein 2/genetics , Methyl-CpG-Binding Protein 2/metabolism , Myotonic Dystrophy/genetics , Myotonic Dystrophy/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Organoids/metabolism , RNA Splicing , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Trinucleotide Repeat Expansion
4.
Brain Res ; 1732: 146683, 2020 04 01.
Article in English | MEDLINE | ID: mdl-32001243

ABSTRACT

Nearly one-hundred loci in the human genome have been associated with different forms of Charcot-Marie-Tooth disease (CMT) and related inherited neuropathies. Despite this wealth of gene targets, treatment options are still extremely limited, and clear "druggable" pathways are not obvious for many of these mutations. However, recent advances in gene therapies are beginning to circumvent this challenge. Each type of CMT is a monogenic disorder, and the cellular targets are usually well-defined and typically include peripheral neurons or Schwann cells. In addition, the genetic mechanism is often also clear, with loss-of-function mutations requiring restoration of gene expression, and gain-of-function or dominant-negative mutations requiring silencing of the mutant allele. These factors combine to make CMT a good target for developing genetic therapies. Here we will review the state of relatively established gene therapy approaches, including viral vector-mediated gene replacement and antisense oligonucleotides for exon skipping, altering splicing, and gene knockdown. We will also describe earlier stage approaches for allele-specific knockdown and CRIPSR/Cas9 gene editing. We will next describe how these various approaches have been deployed in clinical and preclinical studies. Finally, we will evaluate various forms of CMT as candidates for gene therapy based on the current understanding of their genetics, cellular/tissue targets, validated animal models, and availability of patient populations and natural history data.


Subject(s)
Charcot-Marie-Tooth Disease/therapy , Genetic Therapy/methods , Schwann Cells/metabolism , Animals , Charcot-Marie-Tooth Disease/genetics , Charcot-Marie-Tooth Disease/metabolism , Humans
5.
J Clin Invest ; 129(12): 5568-5583, 2019 12 02.
Article in English | MEDLINE | ID: mdl-31557132

ABSTRACT

Gene therapy approaches are being deployed to treat recessive genetic disorders by restoring the expression of mutated genes. However, the feasibility of these approaches for dominantly inherited diseases - where treatment may require reduction in the expression of a toxic mutant protein resulting from a gain-of-function allele - is unclear. Here we show the efficacy of allele-specific RNAi as a potential therapy for Charcot-Marie-Tooth disease type 2D (CMT2D), caused by dominant mutations in glycyl-tRNA synthetase (GARS). A de novo mutation in GARS was identified in a patient with a severe peripheral neuropathy, and a mouse model precisely recreating the mutation was produced. These mice developed a neuropathy by 3-4 weeks of age, validating the pathogenicity of the mutation. RNAi sequences targeting mutant GARS mRNA, but not wild-type, were optimized and then packaged into AAV9 for in vivo delivery. This almost completely prevented the neuropathy in mice treated at birth. Delaying treatment until after disease onset showed modest benefit, though this effect decreased the longer treatment was delayed. These outcomes were reproduced in a second mouse model of CMT2D using a vector specifically targeting that allele. The effects were dose dependent, and persisted for at least 1 year. Our findings demonstrate the feasibility of AAV9-mediated allele-specific knockdown and provide proof of concept for gene therapy approaches for dominant neuromuscular diseases.


Subject(s)
Charcot-Marie-Tooth Disease/therapy , Genetic Therapy , Glycine-tRNA Ligase/genetics , RNA Interference , Alleles , Animals , Disease Models, Animal , HEK293 Cells , Humans , Mice , Mutation
6.
ACS Chem Neurosci ; 10(6): 2890-2902, 2019 06 19.
Article in English | MEDLINE | ID: mdl-31017387

ABSTRACT

Hereditary demyelinating neuropathies linked to peripheral myelin protein 22 (PMP22) involve the disruption of normal protein trafficking and are therefore relevant targets for chaperone therapy. Using a small molecule HSP90 inhibitor, EC137, in cell culture models, we previously validated the chaperone pathway as a viable target for therapy development. Here, we tested five commercially available inhibitors of HSP90 and identified BIIB021 and AUY922 to support Schwann cell viability and enhance chaperone expression. AUY922 showed higher efficacy, compared to BIIB021, in enhancing myelin synthesis in dorsal root ganglion explant cultures from neuropathic mice. For in vivo testing, we randomly assigned 2-3 month old C22 and 6 week old Trembler J (TrJ) mice to receive two weekly injections of either vehicle or AUY922 (2 mg/kg). By the intraperitoneal (i.p.) route, the drug was well-tolerated by all mice over the 5 month long study, without influence on body weight or general grooming behavior. AUY922 improved the maintenance of myelinated nerves of both neuropathic models and attenuated the decline in rotarod performance and peak muscle force production in C22 mice. These studies highlight the significance of proteostasis in neuromuscular function and further validate the HSP90 pathway as a therapeutic target for hereditary neuropathies.


Subject(s)
Charcot-Marie-Tooth Disease/pathology , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Isoxazoles/pharmacology , Nerve Fibers, Myelinated/drug effects , Resorcinols/pharmacology , Animals , Axons/drug effects , Axons/pathology , Mice , Myelin Sheath/drug effects , Myelin Sheath/pathology , Nerve Fibers, Myelinated/pathology
7.
Cell Rep ; 18(13): 3178-3191, 2017 03 28.
Article in English | MEDLINE | ID: mdl-28355569

ABSTRACT

Charcot-Marie-Tooth (CMT) disease is a clinically and genetically heterogeneous group of inherited polyneuropathies. Mutations in 80 genetic loci can cause forms of CMT, resulting in demyelination and axonal dysfunction. The clinical presentation, including sensory deficits, distal muscle weakness, and atrophy, can vary greatly in severity and progression. Here, we used mouse models of CMT to demonstrate genetic interactions that result in a more severe neuropathy phenotype. The cell adhesion molecule Nrcam and the Na+ channel Scn8a (NaV1.6) are important components of nodes. Homozygous Nrcam and heterozygous Scn8a mutations synergized with both an Sh3tc2 mutation, modeling recessive demyelinating Charcot-Marie-Tooth type 4C, and mutations in Gars, modeling dominant axonal Charcot-Marie-Tooth type 2D. We conclude that genetic variants perturbing the structure and function of nodes interact with mutations affecting the cable properties of axons by thinning myelin or reducing axon diameter. Therefore, genes integral to peripheral nodes are candidate modifiers of peripheral neuropathy.


Subject(s)
Charcot-Marie-Tooth Disease/genetics , Demyelinating Diseases/genetics , Peripheral Nerves/pathology , Animals , Axons/metabolism , Carrier Proteins/genetics , Cell Adhesion Molecules/genetics , Charcot-Marie-Tooth Disease/pathology , Demyelinating Diseases/pathology , Disease Models, Animal , Heterozygote , Intracellular Signaling Peptides and Proteins , Mice, Inbred C57BL , Mutation/genetics , NAV1.6 Voltage-Gated Sodium Channel/genetics , Neuromuscular Junction/metabolism
8.
Biol Open ; 5(7): 908-20, 2016 Jul 15.
Article in English | MEDLINE | ID: mdl-27288508

ABSTRACT

Charcot-Marie-Tooth disease encompasses a genetically heterogeneous class of heritable polyneuropathies that result in axonal degeneration in the peripheral nervous system. Charcot-Marie-Tooth type 2D neuropathy (CMT2D) is caused by dominant mutations in glycyl tRNA synthetase (GARS). Mutations in the mouse Gars gene result in a genetically and phenotypically valid animal model of CMT2D. How mutations in GARS lead to peripheral neuropathy remains controversial. To identify putative disease mechanisms, we compared metabolites isolated from the spinal cord of Gars mutant mice and their littermate controls. A profile of altered metabolites that distinguish the affected and unaffected tissue was determined. Ascorbic acid was decreased fourfold in the spinal cord of CMT2D mice, but was not altered in serum. Carnitine and its derivatives were also significantly reduced in spinal cord tissue of mutant mice, whereas glycine was elevated. Dietary supplementation with acetyl-L-carnitine improved gross motor performance of CMT2D mice, but neither acetyl-L-carnitine nor glycine supplementation altered the parameters directly assessing neuropathy. Other metabolite changes suggestive of liver and kidney dysfunction in the CMT2D mice were validated using clinical blood chemistry. These effects were not secondary to the neuromuscular phenotype, as determined by comparison with another, genetically unrelated mouse strain with similar neuromuscular dysfunction. However, these changes do not seem to be causative or consistent metabolites of CMT2D, because they were not observed in a second mouse Gars allele or in serum samples from CMT2D patients. Therefore, the metabolite 'fingerprint' we have identified for CMT2D improves our understanding of cellular biochemical changes associated with GARS mutations, but identification of efficacious treatment strategies and elucidation of the disease mechanism will require additional studies.

9.
J Neurosci ; 36(11): 3254-67, 2016 Mar 16.
Article in English | MEDLINE | ID: mdl-26985035

ABSTRACT

Patients with Charcot-Marie-Tooth Type 2D (CMT2D), caused by dominant mutations in Glycl tRNA synthetase (GARS), present with progressive weakness, consistently in the hands, but often in the feet also. Electromyography shows denervation, and patients often report that early symptoms include cramps brought on by cold or exertion. Based on reported clinical observations, and studies of mouse models of CMT2D, we sought to determine whether weakened synaptic transmission at the neuromuscular junction (NMJ) is an aspect of CMT2D. Quantal analysis of NMJs in two different mouse models of CMT2D (Gars(P278KY), Gars(C201R)), found synaptic deficits that correlated with disease severity and progressed with age. Results of voltage-clamp studies revealed presynaptic defects characterized by: (1) decreased frequency of spontaneous release without any change in quantal amplitude (miniature endplate current), (2) reduced amplitude of evoked release (endplate current) and quantal content, (3) age-dependent changes in the extent of depression in response to repetitive stimulation, and (4) release failures at some NMJs with high-frequency, long-duration stimulation. Drugs that modify synaptic efficacy were tested to see whether neuromuscular performance improved. The presynaptic action of 3,4 diaminopyridine was not beneficial, whereas postsynaptic-acting physostigmine did improve performance. Smaller mutant NMJs with correspondingly fewer vesicles and partial denervation that eliminates some release sites also contribute to the reduction of release at a proportion of mutant NMJs. Together, these voltage-clamp data suggest that a number of release processes, while essentially intact, likely operate suboptimally at most NMJs of CMT2D mice. SIGNIFICANCE STATEMENT: We have uncovered a previously unrecognized aspect of axonal Charcot-Marie-Tooth disease in mouse models of CMT2D. Synaptic dysfunction contributes to impaired neuromuscular performance and disease progression. This suggests that drugs which improve synaptic efficacy at the NMJ could be considered in treating the pathophysiology of CMT2D patients.


Subject(s)
Charcot-Marie-Tooth Disease/pathology , Disease Models, Animal , Glycine-tRNA Ligase/genetics , Mutation/genetics , Neuromuscular Junction/pathology , Synaptic Transmission/genetics , Age Factors , Aminopyridines/pharmacology , Animals , Charcot-Marie-Tooth Disease/genetics , Charcot-Marie-Tooth Disease/physiopathology , Electric Stimulation , Imaging, Three-Dimensional , Mice , Mice, Transgenic , Motor Endplate/genetics , Motor Endplate/physiopathology , Muscle Strength/genetics , Muscle, Skeletal/pathology , Muscle, Skeletal/ultrastructure , Nerve Tissue Proteins/metabolism , Neuromuscular Junction/drug effects , Neuromuscular Junction/genetics , Neuromuscular Junction/metabolism , Patch-Clamp Techniques , Receptors, Cholinergic/metabolism , Synaptic Potentials/drug effects , Synaptic Potentials/genetics , Synaptic Vesicles/pathology , Synaptic Vesicles/ultrastructure
10.
J Neuropathol Exp Neurol ; 73(7): 693-701, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24918641

ABSTRACT

Mutations in HINT1, the gene encoding histidine triad nucleotide-binding protein 1 (HINT1), cause a recessively inherited peripheral neuropathy that primarily involves motor dysfunction and is usually associated with neuromyotonia (i.e. prolonged muscle contraction resulting from hyperexcitability of peripheral nerves). Because these mutations are hypothesized to cause loss of function, we analyzed Hint1 knockout mice for their relevance as a disease model. Mice lacking Hint1 appeared normal and yielded normal behavioral test results or motor performance, although they moved more slowly and for a smaller fraction of time in an open-field arena than wild-type mice. Muscles, neuromuscular junctions, and nodes of Ranvier were anatomically normal and did not show evidence of degeneration or regeneration. Axon numbers and myelination in peripheral nerves were normal at ages 4 and 13 months. Axons were slightly smaller than those in wild-type mice at age 4 months, but this did not cause a decrease in conduction velocity, and no differences in axon diameters were detected at 13 months. With electromyography, we were unable to detect neuromyotonia even after using supraphysiologic stimuli and stressors such as reduced temperature or 3,4-diaminopyridine to block potassium channels. Therefore, we conclude that Hint1 knockout mice may be useful for studying the biochemical activities of HINT1, but these mice do not provide a disease model or a means for investigating the basis of HINT1-associated neuropathy and neuromyotonia.


Subject(s)
Isaacs Syndrome/genetics , Isaacs Syndrome/pathology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/physiology , Peripheral Nervous System Diseases/genetics , Peripheral Nervous System Diseases/pathology , 4-Aminopyridine/analogs & derivatives , Amifampridine , Animals , Anxiety/genetics , Anxiety/psychology , Axons/ultrastructure , Behavior, Animal/physiology , Electromyography , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/physiology , Muscle, Skeletal/innervation , Muscle, Skeletal/pathology , Mutation/physiology , Phenotype , Postural Balance/physiology , Potassium Channel Blockers/pharmacology , Psychomotor Performance/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...