Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Hum Mol Genet ; 33(6): 510-519, 2024 Feb 28.
Article in English | MEDLINE | ID: mdl-38073249

ABSTRACT

Spinal muscular atrophy (SMA) is an autosomal recessive neuromuscular disease characterized by motor neuron loss and skeletal muscle atrophy. SMA is caused by the loss of the SMN1 gene and low SMN protein levels. Current SMA therapies work by increasing SMN protein in the body. Although SMA is regarded as a motor neuron disorder, growing evidence shows that several peripheral organs contribute to SMA pathology. A gene therapy treatment, onasemnogene abeparvovec, is being explored in clinical trials via both systemic and central nervous system (CNS) specific delivery, but the ideal route of delivery as well as the long-term effectiveness is unclear. To investigate the impact of gene therapy long term, we assessed SMA mice at 6 months after treatment of either intravenous (IV) or intracerebroventricular (ICV) delivery of scAAV9-cba-SMN. Interestingly, we observed that SMN protein levels were restored in the peripheral tissues but not in the spinal cord at 6 months of age. However, ICV injections provided better motor neuron and motor function protection than IV injection, while IV-injected mice demonstrated better protection of neuromuscular junctions and muscle fiber size. Surprisingly, both delivery routes resulted in an equal rescue on survival, weight, and liver and pancreatic defects. These results demonstrate that continued peripheral AAV9-SMN gene therapy is beneficial for disease improvement even in the absence of SMN restoration in the spinal cord.


Subject(s)
Muscular Atrophy, Spinal , Animals , Mice , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/therapy , Motor Neurons , Disease Models, Animal , Central Nervous System , Genetic Therapy
2.
Nat Biomed Eng ; 8(2): 118-131, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38057426

ABSTRACT

Spinal muscular atrophy (SMA) is caused by mutations in SMN1. SMN2 is a paralogous gene with a C•G-to-T•A transition in exon 7, which causes this exon to be skipped in most SMN2 transcripts, and results in low levels of the protein survival motor neuron (SMN). Here we show, in fibroblasts derived from patients with SMA and in a mouse model of SMA that, irrespective of the mutations in SMN1, adenosine base editors can be optimized to target the SMN2 exon-7 mutation or nearby regulatory elements to restore the normal expression of SMN. After optimizing and testing more than 100 guide RNAs and base editors, and leveraging Cas9 variants with high editing fidelity that are tolerant of different protospacer-adjacent motifs, we achieved the reversion of the exon-7 mutation via an A•T-to-G•C edit in up to 99% of fibroblasts, with concomitant increases in the levels of the SMN2 exon-7 transcript and of SMN. Targeting the SMN2 exon-7 mutation via base editing or other CRISPR-based methods may provide long-lasting outcomes to patients with SMA.


Subject(s)
Muscular Atrophy, Spinal , RNA-Binding Proteins , Mice , Animals , Humans , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , SMN Complex Proteins/genetics , RNA, Guide, CRISPR-Cas Systems , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/therapy , Exons/genetics , Survival of Motor Neuron 2 Protein/genetics
3.
bioRxiv ; 2023 Jan 21.
Article in English | MEDLINE | ID: mdl-36711797

ABSTRACT

Spinal muscular atrophy (SMA) is a devastating neuromuscular disease caused by mutations in the SMN1 gene. Despite the development of various therapies, outcomes can remain suboptimal in SMA infants and the duration of such therapies are uncertain. SMN2 is a paralogous gene that mainly differs from SMN1 by a C•G-to-T•A transition in exon 7, resulting in the skipping of exon 7 in most SMN2 transcripts and production of only low levels of survival motor neuron (SMN) protein. Genome editing technologies targeted to the SMN2 exon 7 mutation could offer a therapeutic strategy to restore SMN protein expression to normal levels irrespective of the patient SMN1 mutation. Here, we optimized a base editing approach to precisely edit SMN2, reverting the exon 7 mutation via an A•T-to-G•C base edit. We tested a range of different adenosine base editors (ABEs) and Cas9 enzymes, resulting in up to 99% intended editing in SMA patient-derived fibroblasts with concomitant increases in SMN2 exon 7 transcript expression and SMN protein levels. We generated and characterized ABEs fused to high-fidelity Cas9 variants which reduced potential off-target editing. Delivery of these optimized ABEs via dual adeno-associated virus (AAV) vectors resulted in precise SMN2 editing in vivo in an SMA mouse model. This base editing approach to correct SMN2 should provide a long-lasting genetic treatment for SMA with advantages compared to current nucleic acid, small molecule, or exogenous gene replacement therapies. More broadly, our work highlights the potential of PAMless SpRY base editors to install edits efficiently and safely.

4.
Front Cell Neurosci ; 16: 972029, 2022.
Article in English | MEDLINE | ID: mdl-35990890

ABSTRACT

Spinal muscular atrophy (SMA) is a monogenic neuromuscular disease caused by low levels of the Survival Motor Neuron (SMN) protein. Motor neuron degeneration is the central hallmark of the disease. However, the SMN protein is ubiquitously expressed and depletion of the protein in peripheral tissues results in intrinsic disease manifestations, including muscle defects, independent of neurodegeneration. The approved SMN-restoring therapies have led to remarkable clinical improvements in SMA patients. Yet, the presence of a significant number of non-responders stresses the need for complementary therapeutic strategies targeting processes which do not rely solely on restoring SMN. Dysregulated cell death pathways are candidates for SMN-independent pathomechanisms in SMA. Receptor-interacting protein kinase 1 (RIPK1) and RIPK3 have been widely recognized as critical therapeutic targets of necroptosis, an important form of programmed cell death. In addition, Caspase-1 plays a fundamental role in inflammation and cell death. In this study, we evaluate the role of necroptosis, particularly RIPK3 and Caspase-1, in the Smn 2B/- mouse model of SMA. We have generated a triple mutant (TKO), the Smn 2B/-; Ripk3 -/-; Casp1 -/- mouse. TKO mice displayed a robust increase in survival and improved motor function compared to Smn 2B/- mice. While there was no protection against motor neuron loss or neuromuscular junction pathology, larger muscle fibers were observed in TKO mice compared to Smn 2B/- mice. Our study shows that necroptosis modulates survival, motor behavior and muscle fiber size independent of SMN levels and independent of neurodegeneration. Thus, small-molecule inhibitors of necroptosis as a combinatorial approach together with SMN-restoring drugs could be a future strategy for the treatment of SMA.

5.
Gene Ther ; 29(9): 544-554, 2022 09.
Article in English | MEDLINE | ID: mdl-35462564

ABSTRACT

Spinal muscular atrophy (SMA) is a neuromuscular disease caused by loss of the SMN1 gene and low SMN protein levels. Although lower motor neurons are a primary target, there is evidence that peripheral organ defects contribute to SMA. Current SMA gene therapy and clinical trials use a single intravenous bolus of the blood-brain-barrier penetrant scAAV9-cba-SMN by either systemic or central nervous system (CNS) delivery, resulting in impressive amelioration of the clinical phenotype but not a complete cure. The impact of scAAV9-cba-SMN treatment regimens on the CNS as well as on specific peripheral organs is yet to be described in a comparative manner. Therefore, we injected SMA mice with scAAV9-cba-SMN either intravenously (IV) for peripheral SMN restoration or intracerebroventricularly (ICV) for CNS-focused SMN restoration. In our system, ICV injections increased SMN in peripheral organs and the CNS while IV administration increased SMN in peripheral tissues only, largely omitting the CNS. Both treatments rescued several peripheral phenotypes while only ICV injections were neuroprotective. Surprisingly, both delivery routes resulted in a robust rescue effect on survival, weight, and motor function, which in IV-treated mice relied on peripheral SMN restoration but not on targeting the motor neurons. This demonstrates the independent contribution of peripheral organs to SMA pathology and suggests that treatments should not be restricted to motor neurons.


Subject(s)
Dependovirus , Muscular Atrophy, Spinal , Animals , Dependovirus/genetics , Disease Models, Animal , Genetic Therapy/methods , Genetic Vectors/genetics , Mice , Motor Neurons/metabolism , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/pathology , Muscular Atrophy, Spinal/therapy , Survival of Motor Neuron 1 Protein/genetics , Survival of Motor Neuron 1 Protein/metabolism
6.
Cell Mol Gastroenterol Hepatol ; 12(1): 354-377.e3, 2021.
Article in English | MEDLINE | ID: mdl-33545428

ABSTRACT

BACKGROUND & AIMS: Nonalcoholic fatty liver disease (NAFLD) is considered a health epidemic with potential devastating effects on the patients and the healthcare systems. Current preclinical models of NAFLD are invariably imperfect and generally take a long time to develop. A mouse model of survival motor neuron (SMN) depletion (Smn2B/- mice) was recently shown to develop significant hepatic steatosis in less than 2 weeks from birth. The rapid onset of fatty liver in Smn2B/- mice provides an opportunity to identify molecular markers of NAFLD. Here, we investigated whether Smn2B/- mice display typical features of NAFLD/nonalcoholic steatohepatitis (NASH). METHODS: Biochemical, histologic, electron microscopy, proteomic, and high-resolution respirometry were used. RESULTS: The Smn2B/- mice develop microvesicular steatohepatitis within 2 weeks, a feature prevented by AAV9-SMN gene therapy. Although fibrosis is not overtly apparent in histologic sections of the liver, there is molecular evidence of fibrogenesis and presence of stellate cell activation. The consequent liver damage arises from mitochondrial reactive oxygen species production and results in hepatic dysfunction in protein output, complement, coagulation, iron homeostasis, and insulin-like growth factor-1 metabolism. The NAFLD phenotype is likely due to non-esterified fatty acid overload from peripheral lipolysis subsequent to hyperglucagonemia compounded by reduced muscle use and insulin resistance. Despite the low hepatic mitochondrial content, isolated mitochondria show enhanced ß-oxidation, likely as a compensatory response, resulting in the production of reactive oxygen species. In contrast to typical NAFLD/NASH, the Smn2B/- mice lose weight because of their associated neurological condition (spinal muscular atrophy) and develop hypoglycemia. CONCLUSIONS: The Smn2B/- mice represent a good model of microvesicular steatohepatitis. Like other models, it is not representative of the complete NAFLD/NASH spectrum. Nevertheless, it offers a reliable, low-cost, early-onset model that is not dependent on diet to identify molecular players in NAFLD pathogenesis and can serve as one of the very few models of microvesicular steatohepatitis for both adult and pediatric populations.


Subject(s)
Disease Models, Animal , Fatty Liver/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Survival of Motor Neuron 1 Protein/metabolism , Animals , Fatty Liver/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Non-alcoholic Fatty Liver Disease/pathology , Survival of Motor Neuron 1 Protein/genetics
7.
EBioMedicine ; 55: 102750, 2020 May.
Article in English | MEDLINE | ID: mdl-32339936

ABSTRACT

BACKGROUND: Mouse models of mild spinal muscular atrophy (SMA) have been extremely challenging to generate. This paucity of model systems has limited our understanding of pathophysiological events in milder forms of the disease and of the effect of SMN depletion during aging. METHODS: A mild mouse model of SMA, termed Smn2B/-;SMN2+/-, was generated by crossing Smn-/-;SMN2 and Smn2B/2B mice. This new model was characterized using behavioral testing, histology, western blot, muscle-nerve electrophysiology as well as ultrasonography to study classical SMA features and extra-neuronal involvement. FINDINGS: Smn2B/-;SMN2+/- mice have normal survival, mild but sustained motor weakness, denervation and neuronal/neuromuscular junction (NMJ) transmission defects, and neurogenic muscle atrophy that are more prominent in male mice. Increased centrally located nuclei, intrinsic contractile and relaxation muscle defects were also identified in both female and male mice, with some male predominance. There was an absence of extra-neuronal pathology. INTERPRETATION: The Smn2B/-;SMN2+/- mouse provides a model of mild SMA, displaying some hallmark features including reduced weight, sustained motor weakness, electrophysiological transmission deficit, NMJ defects, and muscle atrophy. Early and prominent increase central nucleation and intrinsic electrophysiological deficits demonstrate the potential role played by muscle in SMA disease. The use of this model will allow for the understanding of the most susceptible pathogenic molecular changes in motor neurons and muscles, investigation of the effects of SMN depletion in aging, sex differences and most importantly will provide guidance for the currently aging SMA patients treated with the recently approved genetic therapies. FUNDING: This work was supported by Cure SMA/Families of SMA Canada (grant numbers KOT-1819 and KOT-2021); Muscular Dystrophy Association (USA) (grant number 575466); and Canadian Institutes of Health Research (CIHR) (grant number PJT-156379).


Subject(s)
Aging/genetics , Disease Models, Animal , Muscle, Skeletal/physiopathology , Muscular Atrophy, Spinal/physiopathology , Neuromuscular Junction/physiopathology , Survival of Motor Neuron 1 Protein/genetics , Aging/metabolism , Aging/pathology , Animals , Body Weight , Female , Gene Expression , Gene Knockout Techniques , Longevity/genetics , Male , Mice , Mice, Knockout , Motor Activity , Motor Neurons/cytology , Motor Neurons/metabolism , Muscle, Skeletal/innervation , Muscle, Skeletal/metabolism , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/metabolism , Neuromuscular Junction/metabolism , Sciatic Nerve/metabolism , Sciatic Nerve/physiopathology , Severity of Illness Index , Sex Factors , Survival of Motor Neuron 1 Protein/metabolism , Synaptic Transmission/physiology , Tissue Culture Techniques
8.
J Neuromuscul Dis ; 7(3): 315-322, 2020.
Article in English | MEDLINE | ID: mdl-32333548

ABSTRACT

Spinal muscular atrophy (SMA) is a neuromuscular disorder affecting young children. While pre-clinical models of SMA show small spleens, the same is not true in humans. Here, we show by doppler ultrasonography decreased splenic blood flow in Smn2B/- mice. Further, AAV9-SMN gene therapy does not rescue the distal ear and tail necrosis nor the spleen size in these mice, suggesting that the latter may be linked to a cardiovascular defect. Absence of smaller spleens in human patients is likely due to differences in presentation of defects in SMA between pre-clinical mouse models and human patients, particularly the susceptibility to cardiovascular issues.


Subject(s)
Disease Models, Animal , Muscular Atrophy, Spinal , Regional Blood Flow/physiology , Spleen/blood supply , Animals , Genetic Therapy , Genetic Vectors , Mice , Mice, Transgenic , Muscular Atrophy, Spinal/pathology , Muscular Atrophy, Spinal/physiopathology , Muscular Atrophy, Spinal/therapy , Spleen/diagnostic imaging , Survival of Motor Neuron 2 Protein , Ultrasonography, Doppler
9.
Ann Clin Transl Neurol ; 6(11): 2340-2346, 2019 11.
Article in English | MEDLINE | ID: mdl-31608604

ABSTRACT

Spinal muscular atrophy (SMA) is a neuromuscular disorder leading to paralysis and death. Recent evidence shows increased susceptibility to dyslipidemia and liver steatosis in patients. Here, we provide evidence that low fat diets nearly double survival in Smn2B/- mice, a model for SMA, independent of changes in SMN levels, liver steatosis, or enhanced hepatic functions. Liver damage and ketone levels were reduced, implying a lower reliance on fatty acid oxidation. This preclinical proof of concept study provides grounds for controlled clinical investigation of dietary needs and offers evidence to inform nutritional guidelines specific to SMA.


Subject(s)
Diet, Fat-Restricted , Muscular Atrophy, Spinal , Animals , Disease Models, Animal , Mice , Mice, Inbred C57BL , Muscular Atrophy, Spinal/pathology , Muscular Atrophy, Spinal/physiopathology
10.
Ann Clin Transl Neurol ; 6(8): 1519-1532, 2019 08.
Article in English | MEDLINE | ID: mdl-31402618

ABSTRACT

OBJECTIVE: Spinal muscular atrophy (SMA) is an inherited neuromuscular disorder leading to paralysis and subsequent death in young children. Initially considered a motor neuron disease, extra-neuronal involvement is increasingly recognized. The primary goal of this study was to investigate alterations in lipid metabolism in SMA patients and mouse models of the disease. METHODS: We analyzed clinical data collected from a large cohort of pediatric SMA type I-III patients as well as SMA type I liver necropsy data. In parallel, we performed histology, lipid analysis, and transcript profiling in mouse models of SMA. RESULTS: We identify an increased susceptibility to developing dyslipidemia in a cohort of 72 SMA patients and liver steatosis in pathological samples. Similarly, fatty acid metabolic abnormalities were present in all SMA mouse models studied. Specifically, Smn2B/- mice displayed elevated hepatic triglycerides and dyslipidemia, resembling non-alcoholic fatty liver disease (NAFLD). Interestingly, this phenotype appeared prior to denervation. INTERPRETATION: This work highlights metabolic abnormalities as an important feature of SMA, suggesting implementation of nutritional and screening guidelines in patients, as such defects are likely to increase metabolic distress and cardiovascular risk. This study emphasizes the need for a systemic therapeutic approach to ensure maximal benefits for all SMA patients throughout their life.


Subject(s)
Dyslipidemias/etiology , Fatty Acids/genetics , Fatty Acids/metabolism , Fatty Liver/etiology , Muscular Atrophy, Spinal/complications , Animals , Child , Child, Preschool , Disease Models, Animal , Dyslipidemias/genetics , Dyslipidemias/metabolism , Fatty Liver/genetics , Fatty Liver/metabolism , Female , Humans , Infant , Lipid Metabolism/genetics , Male , Mice , Mice, Transgenic , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/metabolism , Survival of Motor Neuron 1 Protein/genetics , Triglycerides/metabolism
11.
eNeuro ; 6(3)2019.
Article in English | MEDLINE | ID: mdl-31097626

ABSTRACT

External thermosensation is crucial to regulate animal behavior and homeostasis, but the development of the mammalian thermosensory system is not well known. We investigated whether temperature could play a role in the control of movements in a mammalian model born very immature, the opossum (Monodelphis domestica). Like other marsupials, at birth the opossum performs alternate and rhythmic movements with its forelimbs (FLs) to reach a teat where it attaches in order to continue its development. It was shown that FL movements can be induced by mechanical stimulation of the snout in in vitro preparations of newborns consisting of the neuraxis with skin and FLs intact. In the present study, we used puff ejections of cold, neutral (bath temperature) and hot liquid directed toward the snout to induce FL responses in such preparations. Either the responses were visually observed under a microscope or triceps muscle activity was recorded. Cold liquid systematically induced FL movements and triceps contractions, but neutral and hot temperatures were less potent to do so. Sections of the trigeminal nerves and removal of the facial skin diminished responses to cold and nearly abolished those to hot and neutral stimulations. Transient receptor potential melastatin 8 (TRPM8) being the major cold receptor cation channel in adult mammals, we employed immunohistochemistry and reverse transcription-polymerase chain reaction (RT-PCR) to test for its expression, but found that it is not expressed before 13 postnatal days. Overall our results indicate that cold thermosensation exerts a strong influence on motor behaviors in newborn opossums.


Subject(s)
Monodelphis/physiology , Motor Activity , Temperature , Thermosensing/physiology , Animals , Animals, Newborn/physiology , Female , Forelimb/physiology , In Vitro Techniques , Locomotion , Male , Monodelphis/growth & development , TRPM Cation Channels/physiology
12.
BMC Mol Biol ; 18(1): 19, 2017 07 20.
Article in English | MEDLINE | ID: mdl-28728573

ABSTRACT

BACKGROUND: RBM10 is an RNA binding protein involved in message stabilization and alternative splicing regulation. The objective of the research described herein was to identify novel targets of RBM10-regulated splicing. To accomplish this, we downregulated RBM10 in human cell lines, using small interfering RNAs, then monitored alternative splicing, using a reverse transcription-PCR screening platform. RESULTS: RBM10 knockdown (KD) provoked alterations in splicing events in 10-20% of the pre-mRNAs, most of which had not been previously identified as RBM10 targets. Hierarchical clustering of the genes affected by RBM10 KD revealed good conservation of alternative exon inclusion or exclusion across cell lines. Pathway annotation showed RAS signaling to be most affected by RBM10 KD. Of particular interest was the finding that splicing of SMN pre-mRNA, encoding the survival of motor neuron (SMN) protein, was influenced by RBM10 KD. Inhibition of RBM10 resulted in preferential expression of the full-length, exon 7 retaining, SMN transcript in four cancer cell lines and one normal skin fibroblast cell line. SMN protein is expressed from two genes, SMN1 and SMN2, but the SMN1 gene is homozygously disrupted in people with spinal muscular atrophy; as a consequence, all of the SMN that is expressed in people with this disease is from the SMN2 gene. Expression analyses using primary fibroblasts from control, carrier and spinal muscle atrophy donors demonstrated that RBM10 KD resulted in preferential expression of the full-length, exon 7 retaining, SMN2 transcript. At the protein level, upregulation of the full-length SMN2 was also observed. Re-expression of RBM10, in a stable RBM10 KD cancer cell line, correlated with a reversion of the KD effect, demonstrating specificity. CONCLUSION: Our work has not only expanded the number of pre-mRNA targets for RBM10, but identified RBM10 as a novel regulator of SMN2 alternative inclusion.


Subject(s)
RNA Precursors/genetics , RNA Splicing , RNA-Binding Proteins/metabolism , Alternative Splicing , Cell Line , Cluster Analysis , Computational Biology/methods , Exons , Fibroblasts , Gene Expression Profiling , Humans , Reproducibility of Results , Signal Transduction , Survival of Motor Neuron 2 Protein/genetics , ras Proteins/metabolism
13.
Acta Neuropathol Commun ; 3: 55, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26374403

ABSTRACT

INTRODUCTION: The term motor neuron disease encompasses a spectrum of disorders in which motor neurons are the lost. Importantly, while some motor neurons are lost early in disease and others remain intact at disease end-stage. This creates a valuable experimental paradigm to investigate the factors that regulate motor neuron vulnerability. Spinal muscular atrophy is a childhood motor neuron disease caused by mutations or deletions in the SMN1 gene. Here, we have performed transcriptional analysis on differentially vulnerable motor neurons from an intermediate mouse model of Spinal muscular atrophy at a presymptomatic time point. RESULTS: We have characterised two differentially vulnerable populations, differing in the level neuromuscular junction loss. Transcriptional analysis on motor neuron cell bodies revealed that reduced Smn levels correlate with a reduction of transcripts associated with the ribosome, rRNA binding, ubiquitination and oxidative phosphorylation. Furthermore, P53 pathway activation precedes neuromuscular junction loss, suggesting that denervation may be a consequence, rather than a cause of motor neuron death in Spinal muscular atrophy. Finally, increased vulnerability correlates with a decrease in the positive regulation of DNA repair. CONCLUSIONS: This study identifies pathways related to the function of Smn and associated with differential motor unit vulnerability, thus presenting a number of exciting targets for future therapeutic development.


Subject(s)
Gene Expression Regulation/genetics , Motor Neurons/metabolism , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/pathology , Survival of Motor Neuron 1 Protein/genetics , Animals , Asymptomatic Diseases , Benzofurans , Bungarotoxins/metabolism , Cell Death/genetics , Disease Models, Animal , Gene Expression Profiling , Intermediate Filaments/metabolism , Laser Capture Microdissection , Membrane Glycoproteins/metabolism , Mice , Mice, Transgenic , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Nerve Tissue Proteins/metabolism , Neuromuscular Junction/metabolism , Neuromuscular Junction/pathology , Oligonucleotide Array Sequence Analysis , Quinolines , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism , Signal Transduction/genetics , Time Factors
14.
PLoS One ; 9(7): e101225, 2014.
Article in English | MEDLINE | ID: mdl-24984019

ABSTRACT

Spinal muscular atrophy is an autosomal recessive neuromuscular disease characterized by the progressive loss of alpha motor neurons in the spinal cord. Trichostatin A (TSA) is a histone deacetylase inhibitor with beneficial effects in spinal muscular atrophy mouse models that carry the human SMN2 transgene. It is currently unclear whether TSA specifically targets the SMN2 gene or whether other genes respond to TSA and in turn provide neuroprotection in SMA mice. We have taken advantage of the Smn2B/- mouse model that does not harbor the human SMN2 transgene, to test the hypothesis that TSA has its beneficial effects through a non-SMN mediated pathway. TSA increased the median lifespan of Smn2B/- mice from twenty days to eight weeks. As well, there was a significant attenuation of weight loss and improved motor behavior. Pen test and righting reflex both showed significant improvement, and motor neurons in the spinal cord of Smn2B/- mice were protected from degeneration. Both the size and maturity of neuromuscular junctions were significantly improved in TSA treated Smn2B/- mice. Of interest, TSA treatment did not increase the levels of Smn protein in mouse embryonic fibroblasts or myoblasts obtained from the Smn2B/- mice. In addition, no change in the level of Smn transcripts or protein in the brain or spinal cord of TSA-treated SMA model mice was observed. Furthermore, TSA did not increase Smn protein levels in the hind limb muscle, heart, or liver of Smn2B/- mice. We therefore conclude that TSA likely exerts its effects independent of the endogenous mouse Smn gene. As such, identification of the pathways regulated by TSA in the Smn2B/- mice could lead to the development of novel therapeutics for treating SMA.


Subject(s)
Histone Deacetylase Inhibitors/therapeutic use , Hydroxamic Acids/therapeutic use , Muscular Atrophy, Spinal/drug therapy , Neuroprotective Agents/therapeutic use , Survival of Motor Neuron 2 Protein/genetics , Animals , Disease Models, Animal , Humans , Mice , Motor Activity/drug effects , Motor Activity/genetics , Motor Neurons/drug effects , Muscular Atrophy, Spinal/genetics
15.
Nat Commun ; 5: 4063, 2014 Jun 05.
Article in English | MEDLINE | ID: mdl-24898859

ABSTRACT

Developing human muscle contains inter-myofibre progenitors expressing Bmp-receptor 1a (Bmpr1a) and Myf5 that respond to stimulation with Bmp4. Here we ablate Bmpr1a in Myf5- and MyoD-expressing cells in vivo. Mutant mice reveal increased intramuscular fat and reduced myofibre size in selected muscles, or following muscle injury. Myo-endothelial progenitors are the most affected cell type: clonal studies demonstrate that ablation of Bmpr1a in myo-endothelial cells results in decreased myogenic activity, while adipogenic differentiation is significantly increased. Downstream phospho-Smad 1, 5, 8 signalling is also severely decreased in mutant myo-endothelial cells. Lineage tracing of endothelial cells using VE-cadherin(Cre) driver failed to reveal a significant contribution of these cells to developing or injured skeletal muscle. Thus, myo-endothelial progenitors with functioning Bmpr1a signalling demonstrate myogenic potential, but their main function in vivo is to inhibit intramuscular adipogenesis, both through a cell-autonomous and a cell-cell interaction mechanism.


Subject(s)
Adipogenesis/genetics , Bone Morphogenetic Protein Receptors, Type I/genetics , Muscle, Skeletal/metabolism , Stem Cells/metabolism , Animals , Antigens, CD , Cadherins , Endothelial Cells/metabolism , Mice , Mice, Knockout , MyoD Protein/metabolism , Myogenic Regulatory Factor 5/metabolism , Signal Transduction , Smad1 Protein/metabolism , Smad5 Protein/metabolism , Smad8 Protein/metabolism
16.
FASEB J ; 28(7): 2955-69, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24687993

ABSTRACT

Previously, we identified family with sequence similarity 65, member B (Fam65b), as a protein transiently up-regulated during differentiation and fusion of human myogenic cells. Silencing of Fam65b expression results in severe reduction of myogenin expression and consequent lack of myoblast fusion. The molecular function of Fam65b and whether misregulation of its expression could be causative of muscle diseases are unknown. Protein pulldowns were used to identify Fam65b-interacting proteins in differentiating human muscle cells and regenerating muscle tissue. In vitro, human muscle cells were treated with histone-deacetylase (HDAC) inhibitors, and expression of Fam65b and interacting proteins was studied. Nontreated cells were used as controls. In vivo, expression of Fam65b was down-regulated in developing zebrafish to determine the effects on muscle development. Fam65b binds to HDAC6 and dysferlin, the protein mutated in limb girdle muscular dystrophy 2B. The tricomplex Fam65b-HDAC6-dysferlin is transient, and Fam65b expression is necessary for the complex to form. Treatment of myogenic cells with pan-HDAC or HDAC6-specific inhibitors alters Fam65b expression, while dysferlin expression does not change. Inhibition of Fam65b expression in developing zebrafish results in abnormal muscle, with low birefringence, tears at the myosepta, and increased embryo lethality. Fam65b is an essential component of the HDAC6-dysferlin complex. Down-regulation of Fam65b in developing muscle causes changes consistent with muscle disease.-Balasubramanian, A., Kawahara, G., Gupta, V. A., Rozkalne, A., Beauvais, A., Kunkel, L. M., Gussoni, E. Fam65b is important for formation of the HDAC6-dysferlin protein complex during myogenic cell differentiation.


Subject(s)
Cell Differentiation/genetics , Histone Deacetylases/metabolism , Membrane Proteins/metabolism , Muscle Cells/metabolism , Muscle Development/genetics , Muscle Proteins/metabolism , Proteins/genetics , Proteins/metabolism , Amino Acid Sequence , Animals , Cell Adhesion Molecules , Cells, Cultured , Down-Regulation/genetics , Dysferlin , Histone Deacetylase 6 , Histone Deacetylases/genetics , Humans , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Muscle Proteins/genetics , Muscle, Skeletal/metabolism , Muscular Diseases/genetics , Muscular Diseases/metabolism , Protein Binding/genetics , Sequence Alignment , Tubulin/genetics , Tubulin/metabolism , Zebrafish
17.
Hum Mol Genet ; 23(13): 3432-44, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24497575

ABSTRACT

Spinal muscular atrophy (SMA) is characterized by motor neuron loss, caused by mutations or deletions in the ubiquitously expressed survival motor neuron 1 (SMN1) gene. We recently identified a novel role for Smn protein in glucose metabolism and pancreatic development in both an intermediate SMA mouse model (Smn(2B/-)) and type I SMA patients. In the present study, we sought to determine if the observed metabolic and pancreatic defects are SMA-dependent. We employed a line of heterozygous Smn-depleted mice (Smn(+/-)) that lack the hallmark SMA neuromuscular pathology and overt phenotype. At 1 month of age, pancreatic/metabolic function of Smn(+/-)mice is indistinguishable from wild type. However, when metabolically challenged with a high-fat diet, Smn(+/-)mice display abnormal localization of glucagon-producing α-cells within the pancreatic islets and increased hepatic insulin and glucagon sensitivity, through increased p-AKT and p-CREB, respectively. Further, aging results in weight gain, an increased number of insulin-producing ß cells, hyperinsulinemia and increased hepatic glucagon sensitivity in Smn(+/-)mice. Our study uncovers and highlights an important function of Smn protein in pancreatic islet development and glucose metabolism, independent of canonical SMA pathology. These findings suggest that carriers of SMN1 mutations and/or deletions may be at an increased risk of developing pancreatic and glucose metabolism defects, as even small depletions in Smn protein may be a risk factor for diet- and age-dependent development of metabolic disorders.


Subject(s)
Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/pathology , Pancreas/metabolism , Pancreas/pathology , Survival of Motor Neuron 1 Protein/genetics , Animals , Male , Mice , Obesity/genetics , Obesity/metabolism , Phenotype
18.
FEBS J ; 280(23): 6097-113, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24102982

ABSTRACT

Mammalian muscle cell differentiation is a complex process of multiple steps for which many of the factors involved have not yet been defined. In a screen to identify the regulators of myogenic cell fusion, we found that the gene for G-protein coupled receptor 56 (GPR56) was transiently up-regulated during the early fusion of human myoblasts. Human mutations in the gene for GPR56 cause the disease bilateral frontoparietal polymicrogyria; however, the consequences of receptor dysfunction on muscle development have not been explored. Using knockout mice, we defined the role of GPR56 in skeletal muscle. GPR56(-/-) myoblasts have decreased fusion and smaller myotube sizes in culture. In addition, a loss of GPR56 expression in muscle cells results in decreases or delays in the expression of myogenic differentiation 1, myogenin and nuclear factor of activated T-cell (NFAT)c2. Our data suggest that these abnormalities result from decreased GPR56-mediated serum response element and NFAT signalling. Despite these changes, no overt differences in phenotype were identified in the muscle of GPR56 knockout mice, which presented only a mild but statistically significant elevation of serum creatine kinase compared to wild-type. In agreement with these findings, clinical data from 13 bilateral frontoparietal polymicrogyria patients revealed mild serum creatine kinase increase in only two patients. In summary, targeted disruption of GPR56 in mice results in myoblast abnormalities. The absence of a severe muscle phenotype in GPR56 knockout mice and human patients suggests that other factors may compensate for the lack of this G-protein coupled receptor during muscle development and that the motor delay observed in these patients is likely not a result of primary muscle abnormalities.


Subject(s)
Cell Fusion , Malformations of Cortical Development/pathology , Muscle Development/physiology , Myoblasts/cytology , NFATC Transcription Factors/metabolism , Receptors, G-Protein-Coupled/physiology , Serum Response Element/genetics , Animals , Blotting, Western , Cell Communication , Cell Differentiation , Cell Proliferation , Cells, Cultured , Humans , Immunoenzyme Techniques , Luciferases/metabolism , Male , Malformations of Cortical Development/genetics , Malformations of Cortical Development/metabolism , Mice , Mice, Knockout , MyoD Protein/genetics , MyoD Protein/metabolism , Myoblasts/metabolism , Myogenin/genetics , Myogenin/metabolism , NFATC Transcription Factors/antagonists & inhibitors , NFATC Transcription Factors/genetics , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction
19.
Neurobiol Dis ; 49: 57-67, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22960106

ABSTRACT

Spinal muscular atrophy (SMA) is a devastating childhood motor neuron disease caused by mutations and deletions within the survival motor neuron 1 (SMN1) gene. Although other tissues may be involved, motor neurons remain primary pathological targets, with loss of neuromuscular junctions (NMJs) representing an early and significant event in pathogenesis. Although defects in axonal outgrowth and pathfinding have been observed in cell culture and in lower organisms upon Smn depletion, developmental defects in mouse models have been less obvious. Here, we have employed the Smn(2B/-) mouse model to investigate NMJ remodelling during SMA pathology, induced reinnervation, and paralysis. We show that whilst NMJs are capable of remodelling during pathogenesis, there is a marked reduction in paralysis-induced remodelling and in the nerve-directed re-organisation of acetylcholine receptors. This reduction in remodelling potential could not be attributed to a decreased rate of axonal growth. Finally, we have identified a loss of terminal Schwann cells which could contribute to the defects in remodelling/maintenance observed. Our work demonstrates that there are specific defects in NMJ remodelling in an intermediate SMA mouse model, which could contribute to or underlie pathogenesis in SMA. The development of strategies that can promote the remodelling potential of NMJs may therefore be of significant benefit to SMA patients.


Subject(s)
Muscular Atrophy, Spinal/pathology , Muscular Atrophy, Spinal/physiopathology , Neuromuscular Junction/pathology , Neuromuscular Junction/physiopathology , Survival of Motor Neuron 1 Protein/metabolism , Acetylcholine/metabolism , Animals , Axons/pathology , Axons/physiology , Botulinum Toxins, Type A , Disease Models, Animal , Mice , Mice, Inbred C57BL , Nerve Regeneration/physiology , Neuronal Plasticity/physiology , Paralysis/pathology , Paralysis/physiopathology , Schwann Cells/pathology , Schwann Cells/physiology , Survival of Motor Neuron 1 Protein/genetics , Tibial Nerve/injuries , Tibial Nerve/pathology , Tibial Nerve/physiopathology
20.
Ann Neurol ; 72(2): 256-68, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22926856

ABSTRACT

OBJECTIVE: Spinal muscular atrophy (SMA) is the number 1 genetic killer of young children. It is caused by mutation or deletion of the survival motor neuron 1 (SMN1) gene. Although SMA is primarily a motor neuron disease, metabolism abnormalities such as metabolic acidosis, abnormal fatty acid metabolism, hyperlipidemia, and hyperglycemia have been reported in SMA patients. We thus initiated an in-depth analysis of glucose metabolism in SMA. METHODS: Glucose metabolism and pancreas development were investigated in the Smn(2B/-) intermediate SMA mouse model and type I SMA patients. RESULTS: Here, we demonstrate in an SMA mouse model a dramatic cell fate imbalance within pancreatic islets, with a predominance of glucagon-producing α cells at the expense of insulin-producing ß cells. These SMA mice display fasting hyperglycemia, hyperglucagonemia, and glucose resistance. We demonstrate similar abnormalities in pancreatic islets from deceased children with the severe infantile form of SMA in association with supportive evidence of glucose intolerance in at least a subset of such children. INTERPRETATION: Our results indicate that defects in glucose metabolism may play an important contributory role in SMA pathogenesis.


Subject(s)
Blood Glucose/metabolism , Glucose Metabolism Disorders/etiology , Pancreatic Diseases/etiology , Spinal Muscular Atrophies of Childhood/complications , Age Factors , Animals , Animals, Newborn , Apoptosis/genetics , Blood Glucose/genetics , Cell Proliferation , Disease Models, Animal , Glucagon/blood , Humans , In Situ Nick-End Labeling , Insulin/blood , Insulin-Secreting Cells/pathology , Islets of Langerhans/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation/genetics , Pancreatic Diseases/genetics , Spinal Muscular Atrophies of Childhood/genetics , Survival of Motor Neuron 1 Protein/genetics , Survival of Motor Neuron 1 Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...