Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 120(47): e2300308120, 2023 Nov 21.
Article in English | MEDLINE | ID: mdl-37976261

ABSTRACT

Spinal muscular atrophy (SMA), the top genetic cause of infant mortality, is characterized by motor neuron degeneration. Mechanisms underlying SMA pathogenesis remain largely unknown. Here, we report that the activity of cyclin-dependent kinase 5 (Cdk5) and the conversion of its activating subunit p35 to the more potent activator p25 are significantly up-regulated in mouse models and human induced pluripotent stem cell (iPSC) models of SMA. The increase of Cdk5 activity occurs before the onset of SMA phenotypes, suggesting that it may be an initiator of the disease. Importantly, aberrant Cdk5 activation causes mitochondrial defects and motor neuron degeneration, as the genetic knockout of p35 in an SMA mouse model rescues mitochondrial transport and fragmentation defects, and alleviates SMA phenotypes including motor neuron hyperexcitability, loss of excitatory synapses, neuromuscular junction denervation, and motor neuron degeneration. Inhibition of the Cdk5 signaling pathway reduces the degeneration of motor neurons derived from SMA mice and human SMA iPSCs. Altogether, our studies reveal a critical role for the aberrant activation of Cdk5 in SMA pathogenesis and suggest a potential target for therapeutic intervention.


Subject(s)
Induced Pluripotent Stem Cells , Muscular Atrophy, Spinal , Animals , Humans , Mice , Cyclin-Dependent Kinase 5/genetics , Cyclin-Dependent Kinase 5/metabolism , Disease Models, Animal , Induced Pluripotent Stem Cells/metabolism , Motor Neurons/metabolism , Muscular Atrophy, Spinal/metabolism , Nerve Degeneration/pathology , Survival of Motor Neuron 1 Protein/genetics , Survival of Motor Neuron 1 Protein/metabolism
2.
Neuron ; 111(9): 1423-1439.e4, 2023 05 03.
Article in English | MEDLINE | ID: mdl-36863345

ABSTRACT

Reduced survival motor neuron (SMN) protein triggers the motor neuron disease, spinal muscular atrophy (SMA). Restoring SMN prevents disease, but it is not known how neuromuscular function is preserved. We used model mice to map and identify an Hspa8G470R synaptic chaperone variant, which suppressed SMA. Expression of the variant in the severely affected mutant mice increased lifespan >10-fold, improved motor performance, and mitigated neuromuscular pathology. Mechanistically, Hspa8G470R altered SMN2 splicing and simultaneously stimulated formation of a tripartite chaperone complex, critical for synaptic homeostasis, by augmenting its interaction with other complex members. Concomitantly, synaptic vesicular SNARE complex formation, which relies on chaperone activity for sustained neuromuscular synaptic transmission, was found perturbed in SMA mice and patient-derived motor neurons and was restored in modified mutants. Identification of the Hspa8G470R SMA modifier implicates SMN in SNARE complex assembly and casts new light on how deficiency of the ubiquitous protein causes motor neuron disease.


Subject(s)
Muscular Atrophy, Spinal , Animals , Mice , Disease Models, Animal , Motor Neurons/metabolism , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/metabolism , Muscular Atrophy, Spinal/pathology , SNARE Proteins/genetics , SNARE Proteins/metabolism , Survival of Motor Neuron 1 Protein/genetics , Survival of Motor Neuron 1 Protein/metabolism , Synapses/metabolism , Synaptic Transmission , Transcription Factors/metabolism
3.
Int J Mol Sci ; 22(15)2021 Jul 27.
Article in English | MEDLINE | ID: mdl-34360794

ABSTRACT

Spinal muscular atrophy (SMA) is a motor neuron disease caused by insufficient levels of the survival motor neuron (SMN) protein. One of the most prominent pathological characteristics of SMA involves defects of the neuromuscular junction (NMJ), such as denervation and reduced clustering of acetylcholine receptors (AChRs). Recent studies suggest that upregulation of agrin, a crucial NMJ organizer promoting AChR clustering, can improve NMJ innervation and reduce muscle atrophy in the delta7 mouse model of SMA. To test whether the muscle-specific kinase (MuSK), part of the agrin receptor complex, also plays a beneficial role in SMA, we treated the delta7 SMA mice with an agonist antibody to MuSK. MuSK agonist antibody #13, which binds to the NMJ, significantly improved innervation and synaptic efficacy in denervation-vulnerable muscles. MuSK agonist antibody #13 also significantly increased the muscle cross-sectional area and myofiber numbers in these denervation-vulnerable muscles but not in denervation-resistant muscles. Although MuSK agonist antibody #13 did not affect the body weight, our study suggests that preservation of NMJ innervation by the activation of MuSK may serve as a complementary therapy to SMN-enhancing drugs to maximize the therapeutic effectiveness for all types of SMA patients.


Subject(s)
Motor Neurons/enzymology , Muscular Atrophy, Spinal/enzymology , Neuromuscular Junction/enzymology , Receptor Protein-Tyrosine Kinases/metabolism , Animals , Disease Models, Animal , Enzyme Activation , Mice , Mice, Transgenic , Motor Neurons/pathology , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/pathology , Neuromuscular Junction/genetics , Neuromuscular Junction/pathology , Receptor Protein-Tyrosine Kinases/genetics , Survival of Motor Neuron 1 Protein/genetics , Survival of Motor Neuron 1 Protein/metabolism
4.
Hum Mol Genet ; 31(1): 82-96, 2021 12 17.
Article in English | MEDLINE | ID: mdl-34368854

ABSTRACT

Spinal muscular atrophy (SMA) is caused by the loss of the survival motor neuron 1 (SMN1) gene function. The related SMN2 gene partially compensates but produces insufficient levels of SMN protein due to alternative splicing of exon 7. Evrysdi™ (risdiplam), recently approved for the treatment of SMA, and related compounds promote exon 7 inclusion to generate full-length SMN2 mRNA and increase SMN protein levels. SMNΔ7 type I SMA mice survive without treatment for ~17 days. SMN2 mRNA splicing modulators increase survival of SMN∆7 mice with treatment initiated at postnatal day 3 (PND3). To define SMN requirements for adult mice, SMNΔ7 mice were dosed with an SMN2 mRNA splicing modifier from PND3 to PND40, then dosing was stopped. Mice not treated after PND40 showed progressive weight loss, necrosis, and muscle atrophy after ~20 days. Male mice presented a more severe phenotype than female mice. Mice dosed continuously did not show disease symptoms. The estimated half-life of SMN protein is 2 days indicating that the SMA phenotype reappeared after SMN protein levels returned to baseline. Although SMN protein levels decreased with age in mice and SMN protein levels were higher in brain than in muscle, our studies suggest that SMN protein is required throughout the life of the mouse and is especially essential in adult peripheral tissues including muscle. These studies indicate that drugs such as risdiplam will be optimally therapeutic when given as early as possible after diagnosis and potentially will be required for the life of an SMA patient.


Subject(s)
Muscular Atrophy, Spinal , Alternative Splicing , Animals , Disease Models, Animal , Disease Progression , Exons , Female , Humans , Male , Mice , Muscular Atrophy, Spinal/metabolism , RNA Splicing , Survival of Motor Neuron 1 Protein/genetics , Survival of Motor Neuron 1 Protein/metabolism , Survival of Motor Neuron 2 Protein
6.
JCI Insight ; 5(12)2020 06 18.
Article in English | MEDLINE | ID: mdl-32516136

ABSTRACT

Spinal muscular atrophy (SMA) is an inherited neuromuscular disorder caused by reduced expression of the survival motor neuron (SMN) protein. SMN has key functions in multiple RNA pathways, including the biogenesis of small nuclear ribonucleoproteins that are essential components of both major (U2-dependent) and minor (U12-dependent) spliceosomes. Here we investigated the specific contribution of U12 splicing dysfunction to SMA pathology through selective restoration of this RNA pathway in mouse models of varying phenotypic severity. We show that virus-mediated delivery of minor snRNA genes specifically improves select U12 splicing defects induced by SMN deficiency in cultured mammalian cells, as well as in the spinal cord and dorsal root ganglia of SMA mice without increasing SMN expression. This approach resulted in a moderate amelioration of several parameters of the disease phenotype in SMA mice, including survival, weight gain, and motor function. Importantly, minor snRNA gene delivery improved aberrant splicing of the U12 intron-containing gene Stasimon and rescued the severe loss of proprioceptive sensory synapses on SMA motor neurons, which are early signatures of motor circuit dysfunction in mouse models. Taken together, these findings establish the direct contribution of U12 splicing dysfunction to synaptic deafferentation and motor circuit pathology in SMA.


Subject(s)
Motor Neurons/metabolism , Muscular Atrophy, Spinal/genetics , RNA, Small Nuclear/genetics , Synapses/metabolism , Animals , Disease Models, Animal , Mice , Muscular Atrophy, Spinal/pathology , RNA Splicing/genetics , RNA, Messenger/metabolism , RNA, Small Nuclear/metabolism , Spinal Cord/metabolism
7.
J Clin Invest ; 130(3): 1271-1287, 2020 03 02.
Article in English | MEDLINE | ID: mdl-32039917

ABSTRACT

Paucity of the survival motor neuron (SMN) protein triggers the oft-fatal infantile-onset motor neuron disorder, spinal muscular atrophy (SMA). Augmenting the protein is one means of treating SMA and recently led to FDA approval of an intrathecally delivered SMN-enhancing oligonucleotide currently in use. Notwithstanding the advent of this and other therapies for SMA, it is unclear whether the paralysis associated with the disease derives solely from dysfunctional motor neurons that may be efficiently targeted by restricted delivery of SMN-enhancing agents to the nervous system, or stems from broader defects of the motor unit, arguing for systemic SMN repletion. We investigated the disease-contributing effects of low SMN in one relevant peripheral organ - skeletal muscle - by selectively depleting the protein in only this tissue. We found that muscle deprived of SMN was profoundly damaged. Although a disease phenotype was not immediately obvious, persistent low levels of the protein eventually resulted in muscle fiber defects, neuromuscular junction abnormalities, compromised motor performance, and premature death. Importantly, restoring SMN after the onset of muscle pathology reversed disease. Our results provide the most compelling evidence yet for a direct contributing role of muscle in SMA and argue that an optimal therapy for the disease must be designed to treat this aspect of the dysfunctional motor unit.


Subject(s)
Motor Neurons/metabolism , Muscle, Skeletal/metabolism , Muscular Atrophy, Spinal/metabolism , Survival of Motor Neuron 1 Protein/metabolism , Animals , Disease Models, Animal , Mice , Mice, Transgenic , Motor Neurons/pathology , Muscle, Skeletal/pathology , Muscular Atrophy, Spinal/drug therapy , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/pathology , Survival of Motor Neuron 1 Protein/genetics
8.
Cell Stem Cell ; 26(2): 131-133, 2020 02 06.
Article in English | MEDLINE | ID: mdl-32032521

ABSTRACT

Stem cell technology enables the production of three-dimensional organ-like structures, but engineering multi-tissue anatomy has proven difficult. In this issue of Cell Stem Cell, Martins et al. (2020) show that generating a common progenitor cell for posterior spinal cord and muscle enables the formation of functional neuromuscular junctions in single organoids.


Subject(s)
Motor Skills , Organoids , Humans , Neuromuscular Junction , Stem Cells , Tissue Engineering
9.
Sci Rep ; 9(1): 7799, 2019 05 24.
Article in English | MEDLINE | ID: mdl-31127156

ABSTRACT

Spinal muscular atrophy (SMA) is caused by loss-of-function mutations in the survival of motoneuron gene 1 (SMN1). SMA is characterized by motoneuron death, skeletal muscle denervation and atrophy. Disease severity inversely correlates with copy number of a second gene (SMN2), which harbors a splicing defect that causes the production of inadequate levels of functional SMN protein. Small molecules that modify SMN2 splicing towards increased production of functional SMN significantly ameliorate SMA phenotypes in mouse models of severe SMA. At suboptimal doses, splicing modifiers, such as SMN-C1, have served to generate mice that model milder SMA, referred to as pharmacological SMA mice, which survive into early adulthood. Nerve sprouting at endplates, known as terminal sprouting, is key to normal muscle fiber reinnervation following nerve injury and its promotion might mitigate neuromuscular symptoms in mild SMA. Sprouting has been difficult to study in severe SMA mice due to their short lifespan. Here, we show that pharmacological SMA mice are capable of terminal sprouting following reinnervation that is largely SMN-C1 dose-independent, but that they display a reinnervation delay that is critically SMN-C1 dose-dependent. Data also suggest that SMN-C1 can induce by itself a limited terminal sprouting response in SMA and wild-type normally-innervated endplates.


Subject(s)
Muscle, Skeletal/innervation , Muscular Atrophy, Spinal/physiopathology , Neuromuscular Junction/physiopathology , Animals , Disease Models, Animal , Humans , Mice , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Muscular Atrophy, Spinal/chemically induced , Muscular Atrophy, Spinal/pathology , Nerve Regeneration , Neuromuscular Junction/pathology , Schwann Cells/pathology
10.
Am J Hum Genet ; 104(4): 638-650, 2019 04 04.
Article in English | MEDLINE | ID: mdl-30905397

ABSTRACT

Familial dysautonomia (FD) is a recessive neurodegenerative disease caused by a splice mutation in Elongator complex protein 1 (ELP1, also known as IKBKAP); this mutation leads to variable skipping of exon 20 and to a drastic reduction of ELP1 in the nervous system. Clinically, many of the debilitating aspects of the disease are related to a progressive loss of proprioception; this loss leads to severe gait ataxia, spinal deformities, and respiratory insufficiency due to neuromuscular incoordination. There is currently no effective treatment for FD, and the disease is ultimately fatal. The development of a drug that targets the underlying molecular defect provides hope that the drastic peripheral neurodegeneration characteristic of FD can be halted. We demonstrate herein that the FD mouse TgFD9;IkbkapΔ20/flox recapitulates the proprioceptive impairment observed in individuals with FD, and we provide the in vivo evidence that postnatal correction, promoted by the small molecule kinetin, of the mutant ELP1 splicing can rescue neurological phenotypes in FD. Daily administration of kinetin starting at birth improves sensory-motor coordination and prevents the onset of spinal abnormalities by stopping the loss of proprioceptive neurons. These phenotypic improvements correlate with increased amounts of full-length ELP1 mRNA and protein in multiple tissues, including in the peripheral nervous system (PNS). Our results show that postnatal correction of the underlying ELP1 splicing defect can rescue devastating disease phenotypes and is therefore a viable therapeutic approach for persons with FD.


Subject(s)
Dysautonomia, Familial/therapy , Kinetin/therapeutic use , Proprioception , RNA Splicing , Transcriptional Elongation Factors/genetics , Alleles , Animals , Behavior, Animal , Cell Line , Crosses, Genetic , Disease Models, Animal , Dysautonomia, Familial/genetics , Exons , Fibroblasts , Genotype , Humans , Introns , Kinetin/genetics , Male , Mice , Mice, Inbred C57BL , Mutation , Neurons/metabolism , Phenotype
11.
J Med Chem ; 61(15): 6501-6517, 2018 Aug 09.
Article in English | MEDLINE | ID: mdl-30044619

ABSTRACT

SMA is an inherited disease that leads to loss of motor function and ambulation and a reduced life expectancy. We have been working to develop orally administrated, systemically distributed small molecules to increase levels of functional SMN protein. Compound 2 was the first SMN2 splicing modifier tested in clinical trials in healthy volunteers and SMA patients. It was safe and well tolerated and increased SMN protein levels up to 2-fold in patients. Nevertheless, its development was stopped as a precautionary measure because retinal toxicity was observed in cynomolgus monkeys after chronic daily oral dosing (39 weeks) at exposures in excess of those investigated in patients. Herein, we describe the discovery of 1 (risdiplam, RG7916, RO7034067) that focused on thorough pharmacology, DMPK and safety characterization and optimization. This compound is undergoing pivotal clinical trials and is a promising medicine for the treatment of patients in all ages and stages with SMA.


Subject(s)
Azo Compounds/pharmacology , Drug Discovery , Muscular Atrophy, Spinal/drug therapy , Muscular Atrophy, Spinal/genetics , Pyrimidines/pharmacology , RNA Splicing/drug effects , Survival of Motor Neuron 2 Protein/genetics , Animals , Azo Compounds/adverse effects , Azo Compounds/therapeutic use , Humans , Pyrimidines/adverse effects , Pyrimidines/therapeutic use , Safety
12.
Mol Ther ; 24(9): 1592-601, 2016 09.
Article in English | MEDLINE | ID: mdl-27401142

ABSTRACT

Loss of Survival Motor Neuron-1 (SMN1) causes Spinal Muscular Atrophy, a devastating neurodegenerative disease. SMN2 is a nearly identical copy gene; however SMN2 cannot prevent disease development in the absence of SMN1 since the majority of SMN2-derived transcripts are alternatively spliced, encoding a truncated, unstable protein lacking exon 7. Nevertheless, SMN2 retains the ability to produce low levels of functional protein. Previously we have described a splice-switching Morpholino antisense oligonucleotide (ASO) sequence that targets a potent intronic repressor, Element1 (E1), located upstream of SMN2 exon 7. In this study, we have assessed a novel panel of Morpholino ASOs with the goal of optimizing E1 ASO activity. Screening for efficacy in the SMNΔ7 mouse model, a single ASO variant was more active in vivo compared with the original E1(MO)-ASO. Sequence variant eleven (E1(MOv11)) consistently showed greater efficacy by increasing the lifespan of severe Spinal Muscular Atrophy mice after a single intracerebroventricular injection in the central nervous system, exhibited a strong dose-response across an order of magnitude, and demonstrated excellent target engagement by partially reversing the pathogenic SMN2 splicing event. We conclude that Morpholino modified ASOs are effective in modifying SMN2 splicing and have the potential for future Spinal Muscular Atrophy clinical applications.


Subject(s)
Introns , Morpholinos/genetics , Muscular Atrophy, Spinal/genetics , Response Elements , Animals , Disease Models, Animal , Gene Expression Regulation , Gene Targeting , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Mice , Mice, Knockout , Muscular Atrophy, Spinal/metabolism , Muscular Atrophy, Spinal/mortality , Mutation , Prognosis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Survival of Motor Neuron 1 Protein/genetics , Survival of Motor Neuron 1 Protein/metabolism , Transcription, Genetic
13.
Hum Mol Genet ; 25(10): 1885-1899, 2016 05 15.
Article in English | MEDLINE | ID: mdl-26931466

ABSTRACT

Spinal muscular atrophy (SMA) is caused by the loss or mutation of both copies of the survival motor neuron 1 (SMN1) gene. The related SMN2 gene is retained, but due to alternative splicing of exon 7, produces insufficient levels of the SMN protein. Here, we systematically characterize the pharmacokinetic and pharmacodynamics properties of the SMN splicing modifier SMN-C1. SMN-C1 is a low-molecular weight compound that promotes the inclusion of exon 7 and increases production of SMN protein in human cells and in two transgenic mouse models of SMA. Furthermore, increases in SMN protein levels in peripheral blood mononuclear cells and skin correlate with those in the central nervous system (CNS), indicating that a change of these levels in blood or skin can be used as a non-invasive surrogate to monitor increases of SMN protein levels in the CNS. Consistent with restored SMN function, SMN-C1 treatment increases the levels of spliceosomal and U7 small-nuclear RNAs and corrects RNA processing defects induced by SMN deficiency in the spinal cord of SMNΔ7 SMA mice. A 100% or greater increase in SMN protein in the CNS of SMNΔ7 SMA mice robustly improves the phenotype. Importantly, a ∼50% increase in SMN leads to long-term survival, but the SMA phenotype is only partially corrected, indicating that certain SMA disease manifestations may respond to treatment at lower doses. Overall, we provide important insights for the translation of pre-clinical data to the clinic and further therapeutic development of this series of molecules for SMA treatment.


Subject(s)
Isocoumarins/administration & dosage , Muscular Atrophy, Spinal/drug therapy , Muscular Atrophy, Spinal/genetics , Piperazines/administration & dosage , Small Molecule Libraries/pharmacokinetics , Survival of Motor Neuron 2 Protein/genetics , Alternative Splicing/drug effects , Alternative Splicing/genetics , Animals , Central Nervous System/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Exons/genetics , Humans , Leukocytes, Mononuclear/drug effects , Mice , Mice, Transgenic , Muscular Atrophy, Spinal/blood , Muscular Atrophy, Spinal/pathology , RNA Splicing/drug effects , RNA Splicing/genetics , Skin/metabolism , Small Molecule Libraries/administration & dosage , Survival of Motor Neuron 2 Protein/blood
14.
J Neurosci ; 36(8): 2543-53, 2016 Feb 24.
Article in English | MEDLINE | ID: mdl-26911699

ABSTRACT

Spinal muscular atrophy (SMA) is a motoneuron disease caused by loss or mutation in Survival of Motor Neuron 1 (SMN1) gene. Recent studies have shown that selective restoration of SMN protein in astrocytes partially alleviates pathology in an SMA mouse model, suggesting important roles for astrocytes in SMA. Addressing these underlying mechanisms may provide new therapeutic avenues to fight SMA. Using primary cultures of pure motoneurons or astrocytes from SMNΔ7 (SMA) and wild-type (WT) mice, as well as their mixed and matched cocultures, we characterized the contributions of motoneurons, astrocytes, and their interactions to synapse loss in SMA. In pure motoneuron cultures, SMA motoneurons exhibited normal survival but intrinsic defects in synapse formation and synaptic transmission. In pure astrocyte cultures, SMA astrocytes exhibited defects in calcium homeostasis. In motoneuron-astrocyte contact cocultures, synapse formation and synaptic transmission were significantly reduced when either motoneurons, astrocytes or both were from SMA mice compared with those in WT motoneurons cocultured with WT astrocytes. The reduced synaptic activity is unlikely due to changes in motoneuron excitability. This disruption in synapse formation and synaptic transmission by SMN deficiency was not detected in motoneuron-astrocyte noncontact cocultures. Additionally, we observed a downregulation of Ephrin B2 in SMA astrocytes. These findings suggest that there are both cell autonomous and non-cell-autonomous defects in SMA motoneurons and astrocytes. Defects in contact interactions between SMA motoneurons and astrocytes impair synaptogenesis seen in SMA pathology, possibly due to the disruption of the Ephrin B2 pathway.


Subject(s)
Astrocytes/metabolism , Motor Neurons/metabolism , Muscular Atrophy, Spinal/metabolism , Animals , Astrocytes/pathology , Cell Survival/physiology , Cells, Cultured , Coculture Techniques , Female , Male , Mice , Mice, Transgenic , Motor Neurons/pathology , Muscular Atrophy, Spinal/pathology , Spinal Cord/cytology , Spinal Cord/metabolism , Spinal Cord/pathology
15.
Mol Ther ; 24(5): 855-66, 2016 05.
Article in English | MEDLINE | ID: mdl-26860981

ABSTRACT

Spinal muscular atrophy with respiratory distress type 1 (SMARD1) is an autosomal recessive disease occurring during childhood. The gene responsible for disease development is a ubiquitously expressed protein, IGHMBP2. Mutations in IGHMBP2 result in the loss of α-motor neurons leading to muscle atrophy in the distal limbs accompanied by respiratory complications. Although genetically and clinically distinct, proximal SMA is also caused by the loss of a ubiquitously expressed gene (SMN). Significant preclinical success has been achieved in proximal SMA using viral-based gene replacement strategies. We leveraged the technologies employed in SMA to demonstrate gene replacement efficacy in an SMARD1 animal model. Intracerebroventricular (ICV) injection of single-stranded AAV9 expressing the full-length cDNA of IGHMBP2 in a low dose led to a significant level of rescue in treated SMARD1 animals. Consistent with drastically increased survival, weight gain, and strength, the rescued animals demonstrated a significant improvement in muscle, NMJ, motor neurons, and axonal pathology. In addition, increased levels of IGHMBP2 in lumbar motor neurons verified the efficacy of the virus to transduce the target tissues. Our results indicate that AAV9-based gene replacement is a viable strategy for SMARD1, although dosing effects and potential negative impacts of high dose and ICV injection should be thoroughly investigated.


Subject(s)
DNA-Binding Proteins/genetics , Genetic Therapy , Genetic Vectors/administration & dosage , Muscular Atrophy, Spinal/therapy , Respiratory Distress Syndrome, Newborn/therapy , Transcription Factors/genetics , Animals , Body Weight , Dependovirus/genetics , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Humans , Male , Mice , Muscular Atrophy, Spinal/genetics , Mutation , Respiratory Distress Syndrome, Newborn/genetics , Survival Analysis
16.
Hum Mol Genet ; 25(5): 964-75, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26758873

ABSTRACT

Spinal muscular atrophy (SMA) is a genetic disease characterized by atrophy of muscle and loss of spinal motor neurons. SMA is caused by deletion or mutation of the survival motor neuron 1 (SMN1) gene, and the nearly identical SMN2 gene fails to generate adequate levels of functional SMN protein due to a splicing defect. Currently, several therapeutics targeted to increase SMN protein are in clinical trials. An outstanding issue in the field is whether initiating treatment in symptomatic older patients would confer a therapeutic benefit, an important consideration as the majority of patients with milder forms of SMA are diagnosed at an older age. An SMA mouse model that recapitulates the disease phenotype observed in adolescent and adult SMA patients is needed to address this important question. We demonstrate here that Δ7 mice, a model of severe SMA, treated with a suboptimal dose of an SMN2 splicing modifier show increased SMN protein, survive into adulthood and display SMA disease-relevant pathologies. Increasing the dose of the splicing modifier after the disease symptoms are apparent further mitigates SMA histopathological features in suboptimally dosed adult Δ7 mice. In addition, inhibiting myostatin using intramuscular injection of AAV1-follistatin ameliorates muscle atrophy in suboptimally dosed Δ7 mice. Taken together, we have developed a new murine model of symptomatic SMA in adolescents and adult mice that is induced pharmacologically from a more severe model and demonstrated efficacy of both SMN2 splicing modifiers and a myostatin inhibitor in mice at later disease stages.


Subject(s)
Follistatin/pharmacology , Immunologic Factors/pharmacology , Muscular Atrophy, Spinal/drug therapy , RNA Splicing/drug effects , Survival of Motor Neuron 1 Protein/genetics , Survival of Motor Neuron 2 Protein/agonists , Adolescent , Adult , Age of Onset , Animals , Dependovirus/genetics , Dependovirus/metabolism , Disease Models, Animal , Gene Deletion , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Humans , Mice , Motor Neurons/drug effects , Motor Neurons/metabolism , Motor Neurons/pathology , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/metabolism , Muscular Atrophy, Spinal/pathology , Myostatin/antagonists & inhibitors , Myostatin/genetics , Myostatin/metabolism , Phenotype , Survival of Motor Neuron 1 Protein/metabolism , Survival of Motor Neuron 2 Protein/genetics , Survival of Motor Neuron 2 Protein/metabolism
17.
Cold Spring Harb Perspect Biol ; 7(10): a020503, 2015 Aug 20.
Article in English | MEDLINE | ID: mdl-26430218

ABSTRACT

The neuromuscular junction (NMJ) is engineered to be a highly reliable synapse to carry the control of the motor commands of the nervous system over the muscles. Its development, organization, and synaptic properties are highly structured and regulated to support such reliability and efficacy. Yet, the NMJ is also highly plastic, able to react to injury and adapt to changes. This balance between structural stability and synaptic efficacy on one hand and structural plasticity and repair on another hand is made possible by the intricate regulation of perisynaptic Schwann cells, glial cells at this synapse. They regulate both the efficacy and structural plasticity of the NMJ in a dynamic, bidirectional manner owing to their ability to decode synaptic transmission and by their interactions via trophic-related factors.


Subject(s)
Neuroglia/physiology , Neuromuscular Junction/physiology , Neuronal Plasticity/physiology , Schwann Cells/physiology , Synapses/physiology , Synaptic Transmission/physiology , Animals , Green Fluorescent Proteins/metabolism , Mice , Mice, Transgenic , Microscopy, Fluorescence , Nerve Regeneration , Neurodegenerative Diseases , Xenopus
18.
J Neurosci ; 35(15): 6038-50, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25878277

ABSTRACT

Mechanisms underlying motor neuron degeneration in spinal muscular atrophy (SMA), the leading inherited cause of infant mortality, remain largely unknown. Many studies have established the importance of hyperphosphorylation of the microtubule-associated protein tau in various neurodegenerative disorders, including Alzheimer's and Parkinson's diseases. However, tau phosphorylation in SMA pathogenesis has yet to be investigated. Here we show that tau phosphorylation on serine 202 (S202) and threonine 205 (T205) is increased significantly in SMA motor neurons using two SMA mouse models and human SMA patient spinal cord samples. Interestingly, phosphorylated tau does not form aggregates in motor neurons or neuromuscular junctions (NMJs), even at late stages of SMA disease, distinguishing it from other tauopathies. Hyperphosphorylation of tau on S202 and T205 is mediated by cyclin-dependent kinase 5 (Cdk5) in SMA disease condition, because tau phosphorylation at these sites is significantly reduced in Cdk5 knock-out mice; genetic knock-out of Cdk5 activating subunit p35 in an SMA mouse model also leads to reduced tau phosphorylation on S202 and T205 in the SMA;p35(-/-) compound mutant mice. In addition, expression of the phosphorylation-deficient tauS202A,T205A mutant alleviates motor neuron defects in a zebrafish SMA model in vivo and mouse motor neuron degeneration in culture, whereas expression of phosphorylation-mimetic tauS202E,T205E promotes motor neuron defects. More importantly, genetic knock-out of tau in SMA mice rescues synapse stripping on motor neurons, NMJ denervation, and motor neuron degeneration in vivo. Altogether, our findings suggest a novel mechanism for SMA pathogenesis in which hyperphosphorylation of non-aggregating tau by Cdk5 contributes to motor neuron degeneration.


Subject(s)
Cyclin-Dependent Kinase 5/metabolism , Motor Neurons/pathology , Muscular Atrophy, Spinal , Nerve Degeneration/etiology , Spinal Cord/pathology , tau Proteins/metabolism , Animals , Cells, Cultured , Disease Models, Animal , Female , Gene Expression Regulation/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Homeodomain Proteins/metabolism , Humans , Immunoprecipitation , Infant , Infant, Newborn , Male , Mice , Mice, Transgenic , Motor Neurons/metabolism , Muscle, Skeletal/pathology , Muscular Atrophy, Spinal/complications , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/pathology , Nerve Tissue Proteins/metabolism , Neuromuscular Junction/metabolism , Neuromuscular Junction/pathology , Nuclear Proteins/metabolism , Oligodeoxyribonucleotides, Antisense/pharmacology , Phosphorylation , Repressor Proteins/metabolism , Survival of Motor Neuron 1 Protein/genetics , Survival of Motor Neuron 1 Protein/metabolism , Zebrafish , tau Proteins/deficiency , tau Proteins/genetics
19.
Hum Mol Genet ; 24(14): 4094-102, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-25911676

ABSTRACT

Systemically low levels of survival motor neuron-1 (SMN1) protein cause spinal muscular atrophy (SMA). α-Motor neurons of the spinal cord are considered particularly vulnerable in this genetic disorder and their dysfunction and loss cause progressive muscle weakness, paralysis and eventually premature death of afflicted individuals. Historically, SMA was therefore considered a motor neuron-autonomous disease. However, depletion of SMN in motor neurons of normal mice elicited only a very mild phenotype. Conversely, restoration of SMN to motor neurons in an SMA mouse model had only modest effects on the SMA phenotype and survival. Collectively, these results suggested that additional cell types contribute to the pathogenesis of SMA, and understanding the non-autonomous requirements is crucial for developing effective therapies. Astrocytes are critical for regulating synapse formation and function as well as metabolic support for neurons. We hypothesized that astrocyte functions are disrupted in SMA, exacerbating disease progression. Using viral-based restoration of SMN specifically to astrocytes, survival in severe and intermediate SMA mice was observed. In addition, neuromuscular circuitry was improved. Astrogliosis was prominent in end-stage SMA mice and in post-mortem patient spinal cords. Increased expression of proinflammatory cytokines was partially normalized in treated mice, suggesting that astrocytes contribute to the pathogenesis of SMA.


Subject(s)
Astrocytes/cytology , Astrocytes/metabolism , Muscular Atrophy, Spinal/pathology , Animals , Cell Differentiation , Dependovirus/genetics , Disease Models, Animal , Gene Expression Regulation , Genetic Vectors , Humans , Induced Pluripotent Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Neurons/metabolism , Muscular Atrophy, Spinal/genetics , Neuromuscular Junction/genetics , Neuromuscular Junction/metabolism , Phenotype , Spinal Cord/metabolism , Survival of Motor Neuron 1 Protein/genetics , Survival of Motor Neuron 1 Protein/metabolism
20.
Science ; 345(6197): 688-93, 2014 Aug 08.
Article in English | MEDLINE | ID: mdl-25104390

ABSTRACT

Spinal muscular atrophy (SMA) is a genetic disease caused by mutation or deletion of the survival of motor neuron 1 (SMN1) gene. A paralogous gene in humans, SMN2, produces low, insufficient levels of functional SMN protein due to alternative splicing that truncates the transcript. The decreased levels of SMN protein lead to progressive neuromuscular degeneration and high rates of mortality. Through chemical screening and optimization, we identified orally available small molecules that shift the balance of SMN2 splicing toward the production of full-length SMN2 messenger RNA with high selectivity. Administration of these compounds to Δ7 mice, a model of severe SMA, led to an increase in SMN protein levels, improvement of motor function, and protection of the neuromuscular circuit. These compounds also extended the life span of the mice. Selective SMN2 splicing modifiers may have therapeutic potential for patients with SMA.


Subject(s)
Alternative Splicing/drug effects , Coumarins/administration & dosage , Isocoumarins/administration & dosage , Longevity/drug effects , Muscular Atrophy, Spinal/drug therapy , Pyrimidinones/administration & dosage , Small Molecule Libraries/administration & dosage , Survival of Motor Neuron 2 Protein/genetics , Administration, Oral , Animals , Cells, Cultured , Coumarins/chemistry , Disease Models, Animal , Drug Evaluation, Preclinical , Humans , Isocoumarins/chemistry , Mice , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/metabolism , Pyrimidinones/chemistry , RNA, Messenger/genetics , Sequence Deletion , Small Molecule Libraries/chemistry , Survival of Motor Neuron 2 Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...