Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Hum Mol Genet ; 31(18): 3107-3119, 2022 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-35551393

RESUMEN

Spinal muscular atrophy (SMA) is a childhood motor neuron disease caused by anomalies in the SMN1 gene. Although therapeutics have been approved for the treatment of SMA, there is a therapeutic time window, after which efficacy is reduced. Hallmarks of motor unit pathology in SMA include loss of motor-neurons and neuromuscular junction (NMJs). Following an increase in Smn levels, it is unclear how much damage can be repaired and the degree to which normal connections are re-established. Here, we perform a detailed analysis of motor unit pathology before and after restoration of Smn levels. Using a Smn-inducible mouse model of SMA, we show that genetic restoration of Smn results in a dramatic reduction in NMJ pathology, with restoration of innervation patterns, preservation of axon and endplate number and normalized expression of P53-associated transcripts. Notably, presynaptic swelling and elevated Pmaip levels remained. We analysed the effect of either early or delayed treated of an antisense oligonucleotide (ASO) targeting SMN2 on a range of differentially vulnerable muscles. Following ASO administration, the majority of endplates appeared fully occupied. However, there was an underlying loss of axons and endplates, which was more prevalent following a delay in treatment. There was an increase in average motor unit size following both early and delayed treatment. Together this work demonstrates the remarkably regenerative capacity of the motor neuron following Smn restoration, but highlights that recovery is incomplete. This work suggests that there is an opportunity to enhance neuromuscular junction recovery following administration of Smn-enhancing therapeutics.


Asunto(s)
Atrofia Muscular Espinal , Proteína p53 Supresora de Tumor , Animales , Modelos Animales de Enfermedad , Ratones , Neuronas Motoras/metabolismo , Atrofia Muscular Espinal/genética , Atrofia Muscular Espinal/metabolismo , Atrofia Muscular Espinal/terapia , Oligonucleótidos/farmacología , Oligonucleótidos Antisentido/farmacología , Proteína 1 para la Supervivencia de la Neurona Motora/genética , Proteína 1 para la Supervivencia de la Neurona Motora/metabolismo , Proteína p53 Supresora de Tumor/metabolismo
2.
PLoS Genet ; 13(3): e1006680, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28362802

RESUMEN

The term "motor neuron disease" encompasses a spectrum of disorders in which motor neurons are the primary pathological target. However, in both patients and animal models of these diseases, not all motor neurons are equally vulnerable, in that while some motor neurons are lost very early in disease, others remain comparatively intact, even at late stages. This creates a valuable system to investigate the factors that regulate motor neuron vulnerability. In this study, we aim to use this experimental paradigm to identify potential transcriptional modifiers. We have compared the transcriptome of motor neurons from healthy wild-type mice, which are differentially vulnerable in the childhood motor neuron disease Spinal Muscular Atrophy (SMA), and have identified 910 transcriptional changes. We have compared this data set with published microarray data sets on other differentially vulnerable motor neurons. These neurons were differentially vulnerable in the adult onset motor neuron disease Amyotrophic Lateral Sclerosis (ALS), but the screen was performed on the equivalent population of neurons from neurologically normal human, rat and mouse. This cross species comparison has generated a refined list of differentially expressed genes, including CELF5, Col5a2, PGEMN1, SNCA, Stmn1 and HOXa5, alongside a further enrichment for synaptic and axonal transcripts. As an in vivo validation, we demonstrate that the manipulation of a significant number of these transcripts can modify the neurodegenerative phenotype observed in a Drosophila line carrying an ALS causing mutation. Finally, we demonstrate that vector-mediated expression of alpha-synuclein (SNCA), a transcript decreased in selectively vulnerable motor neurons in all four screens, can extend life span, increase weight and decrease neuromuscular junction pathology in a mouse model of SMA. In summary, we have combined multiple data sets to identify transcripts, which are strong candidates for being phenotypic modifiers, and demonstrated SNCA is a modifier of pathology in motor neuron disease.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Enfermedad de la Neurona Motora/genética , Neuronas Motoras/metabolismo , alfa-Sinucleína/genética , Esclerosis Amiotrófica Lateral/patología , Animales , Axones/metabolismo , Axones/patología , Modelos Animales de Enfermedad , Drosophila melanogaster/genética , Regulación de la Expresión Génica/genética , Humanos , Ratones , Enfermedad de la Neurona Motora/patología , Neuronas Motoras/patología , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Unión Neuromuscular/genética , Unión Neuromuscular/patología , Fenotipo , Ratas , Transcriptoma/genética , alfa-Sinucleína/biosíntesis
3.
Mol Ther ; 24(9): 1592-601, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27401142

RESUMEN

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.


Asunto(s)
Intrones , Morfolinos/genética , Atrofia Muscular Espinal/genética , Elementos de Respuesta , Animales , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Marcación de Gen , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Ratones , Ratones Noqueados , Atrofia Muscular Espinal/metabolismo , Atrofia Muscular Espinal/mortalidad , Mutación , Pronóstico , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteína 1 para la Supervivencia de la Neurona Motora/genética , Proteína 1 para la Supervivencia de la Neurona Motora/metabolismo , Transcripción Genética
4.
Mol Ther ; 24(5): 855-66, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26860981

RESUMEN

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.


Asunto(s)
Proteínas de Unión al ADN/genética , Terapia Genética , Vectores Genéticos/administración & dosificación , Atrofia Muscular Espinal/terapia , Síndrome de Dificultad Respiratoria del Recién Nacido/terapia , Factores de Transcripción/genética , Animales , Peso Corporal , Dependovirus/genética , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Masculino , Ratones , Atrofia Muscular Espinal/genética , Mutación , Síndrome de Dificultad Respiratoria del Recién Nacido/genética , Análisis de Supervivencia
5.
Hum Mol Genet ; 23(17): 4559-68, 2014 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-24722206

RESUMEN

Spinal muscular atrophy (SMA) is a neurodegenerative disease caused by the loss of a single gene, Survival Motor Neuron-1 (SMN1). Administration of a self-complementary Adeno-Associated Virus vector expressing full-length SMN cDNA (scAAV-SMN) has proven an effective means to rescue the SMA phenotype in SMA mice, either by intravenous (IV) or intracerebroventricular (ICV) administration at very early time points. We have recently shown that ICV delivery of scAAV9-SMN is more effective than a similar dose of vector administered via an IV injection, thereby providing an important mechanism to examine a timeline for rescuing the disease and determining the therapeutic window in a severe model of SMA. In this report, we utilized a relatively severe mouse model of SMA, SMNΔ7. Animals were injected with scAAV9-SMN vector via ICV injection on a single day, from P2 through P8. At each delivery point from P2 through P8, scAAV9-SMN decreased disease severity. A near complete rescue was obtained following P2 injection while a P8 injection produced a ∼ 40% extension in survival. Analysis of the underlying neuromuscular junction (NMJ) pathology revealed that late-stage delivery of the vector failed to provide protection from NMJ defects despite robust SMN expression in the central nervous system. While our study demonstrates that a maximal benefit is obtained when treatment is delivered during pre-symptomatic stages, significant therapeutic benefit can still be achieved after the onset of disease symptoms.


Asunto(s)
Atrofia Muscular Espinal/terapia , Animales , Peso Corporal , Pollos , Dependovirus/genética , Modelos Animales de Enfermedad , Terapia Genética , Vectores Genéticos/administración & dosificación , Células HEK293 , Humanos , Inyecciones Intraventriculares , Ratones , Actividad Motora , Músculo Esquelético/patología , Músculo Esquelético/fisiopatología , Atrofia Muscular Espinal/patología , Atrofia Muscular Espinal/fisiopatología , Unión Neuromuscular/patología , Unión Neuromuscular/fisiopatología , Fenotipo , Análisis de Supervivencia , Proteína 1 para la Supervivencia de la Neurona Motora/genética , Proteína 1 para la Supervivencia de la Neurona Motora/uso terapéutico , Factores de Tiempo
6.
Hum Mol Genet ; 23(18): 4832-45, 2014 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-24781211

RESUMEN

Spinal muscular atrophy (SMA) is a neurodegenerative disease caused by the loss of Survival Motor Neuron-1 (SMN1). In all SMA patients, a nearly identical copy gene called SMN2 is present, which produces low levels of functional protein owing to an alternative splicing event. To prevent exon-skipping, we have targeted an intronic repressor, Element1 (E1), located upstream of SMN2 exon 7 using Morpholino-based antisense oligonucleotides (E1(MO)-ASOs). A single intracerebroventricular injection in the relatively severe mouse model of SMA (SMNΔ7 mouse model) elicited a robust induction of SMN protein, and mean life span was extended from an average survival of 13 to 54 days following a single dose, consistent with large weight gains and a correction of the neuronal pathology. Additionally, E1(MO)-ASO treatment in an intermediate SMA mouse (SMN(RT) mouse model) significantly extended life span by ∼700% and weight gain was comparable with the unaffected animals. While a number of experimental therapeutics have targeted the ISS-N1 element of SMN2 pre-mRNA, the development of E1 ASOs provides a new molecular target for SMA therapeutics that dramatically extends survival in two important pre-clinical models of disease.


Asunto(s)
Terapia Genética/métodos , Morfolinos/administración & dosificación , Atrofia Muscular Espinal/terapia , Secuencias Reguladoras de Ácidos Nucleicos , Proteína 2 para la Supervivencia de la Neurona Motora/metabolismo , Animales , Modelos Animales de Enfermedad , Humanos , Intrones , Ratones , Atrofia Muscular Espinal/genética , Atrofia Muscular Espinal/patología , Tasa de Supervivencia , Proteína 2 para la Supervivencia de la Neurona Motora/genética , Aumento de Peso
7.
Biochem Biophys Res Commun ; 470(1): 82-87, 2016 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-26748185

RESUMEN

Spinal Muscular Atrophy (SMA) is an autosomal recessive disorder, leading to fatal loss of motor neurons. It is caused by loss of function of the SMN gene, which is expressed throughout the body, and there is increasing evidence of dysfunction in non-neuronal tissues. Birthweight is one of most powerful prognostic factors for infants born with SMA, and intrauterine growth restriction is common. In the SMNΔ7 mouse model of SMA, pups with the disease lived 25% longer when their mothers were fed a higher fat, "breeder" diet. The placenta is responsible for transport of nutrients from mother to fetus, and is a major determinant of fetal growth. Thus, the present study tested the hypothesis that placental development is impaired in SMNΔ7 conceptuses. Detailed morphological characterization revealed no defects in SMNΔ7 placental development, and expression of key transcription factors regulating mouse placental development was unaffected. The intrauterine growth restriction observed in SMA infants likely does not result from impaired placental development.


Asunto(s)
Atrofia Muscular Espinal/patología , Atrofia Muscular Espinal/fisiopatología , Placentación , Proteínas del Complejo SMN/metabolismo , Trofoblastos/metabolismo , Trofoblastos/patología , Animales , Femenino , Masculino , Ratones , Embarazo
8.
Hum Mol Genet ; 22(9): 1843-55, 2013 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-23390132

RESUMEN

Spinal Muscular Atrophy (SMA) is due to the loss of the survival motor neuron gene 1 (SMN1), resulting in motor neuron (MN) degeneration, muscle atrophy and loss of motor function. While SMN2 encodes a protein identical to SMN1, a single nucleotide difference in exon 7 causes most of the SMN2-derived transcripts to be alternatively spliced resulting in a truncated and unstable protein (SMNΔ7). SMA patients retain at least one SMN2 copy, making it an important target for therapeutics. Many of the existing SMA models are very severe, with animals typically living less than 2 weeks. Here, we present a novel intermediate mouse model of SMA based upon the human genomic SMN2 gene. Genetically, this model is similar to the well-characterized SMNΔ7 model; however, we have manipulated the SMNΔ7 transgene to encode a modestly more functional protein referred to as SMN read-through (SMN(RT)). By introducing the SMN(RT) transgene onto the background of a severe mouse model of SMA (SMN2(+/+);Smn(-/-)), disease severity was significantly decreased based upon a battery of phenotypic parameters, including MN pathology and a significant extension in survival. Importantly, there is not a full phenotypic correction, allowing for the examination of a broad range of therapeutics, including SMN2-dependent and SMN-independent pathways. This novel animal model serves as an important biological and therapeutic model for less severe forms of SMA and provides an in vivo validation of the SMN(RT) protein.


Asunto(s)
Modelos Animales de Enfermedad , Atrofia Muscular Espinal/genética , Proteína 2 para la Supervivencia de la Neurona Motora/genética , Animales , Peso Corporal , Encéfalo/metabolismo , Exones , Regulación de la Expresión Génica , Humanos , Longevidad , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Atrofia Muscular Espinal/patología , Fenotipo , Regiones Promotoras Genéticas , ARN/genética , Empalme del ARN , Médula Espinal/metabolismo , Proteína 1 para la Supervivencia de la Neurona Motora/genética
9.
Mol Ther ; 20(1): 119-26, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22031236

RESUMEN

Spinal muscular atrophy (SMA) is a neurodegenerative disease caused by loss of survival motor neuron-1 (SMN1). A nearly identical copy gene, SMN2, is present in all SMA patients. Although the SMN2 coding sequence has the potential to produce full-length SMN, nearly 90% of SMN2-derived transcripts are alternatively spliced and encode a truncated protein. SMN2, however, is an excellent therapeutic target. Previously, we developed antisense-based oligonucleotides (bifunctional RNAs) that specifically recruit SR/SR-like splicing factors and target a negative regulator of SMN2 exon-7 inclusion within intron-6. As a means to optimize the antisense sequence of the bifunctional RNAs, we chose to target a potent intronic repressor downstream of SMN2 exon 7, called intronic splicing silencer N1 (ISS-N1). We developed RNAs that specifically target ISS-N1 and concurrently recruit the modular SR proteins SF2/ASF or hTra2ß1. RNAs were directly injected in the brains of SMA mice. Bifunctional RNA injections were able to elicit robust induction of SMN protein in the brain and spinal column of neonatal SMA mice. Importantly, hTra2ß1-ISS-N1 and SF2/ASF-ISS-N1 bifunctional RNAs significantly extended lifespan and increased weight in the SMNΔ7 mice. This technology has direct implications for SMA therapy and provides similar therapeutic strategies for other diseases caused by aberrant splicing.


Asunto(s)
Intrones , Atrofia Muscular Espinal/terapia , Oligorribonucleótidos Antisentido/administración & dosificación , Empalme del ARN , Secuencias Reguladoras de Ácido Ribonucleico , Proteínas del Complejo SMN/genética , Animales , Secuencia de Bases , Modelos Animales de Enfermedad , Orden Génico , Marcación de Gen , Terapia Genética , Ratones , Ratones Noqueados , Actividad Motora/genética , Atrofia Muscular Espinal/genética , Atrofia Muscular Espinal/mortalidad , Proteínas del Complejo SMN/metabolismo , Aumento de Peso/genética
10.
Hum Mol Genet ; 18(9): 1600-11, 2009 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-19228773

RESUMEN

Spinal muscular atrophy (SMA) is a motor neuron disease caused by the loss of survival motor neuron-1 (SMN1). A nearly identical copy gene, SMN2, is present in all SMA patients, which produces low levels of functional protein. Although the SMN2 coding sequence has the potential to produce normal, full-length SMN, approximately 90% of SMN2-derived transcripts are alternatively spliced and encode a truncated protein lacking the final coding exon (exon 7). SMN2, however, is an excellent therapeutic target. Previously, we developed bifunctional RNAs that bound SMN exon 7 and modulated SMN2 splicing. To optimize the efficiency of the bifunctional RNAs, a different antisense target was required. To this end, we genetically verified the identity of a putative intronic repressor and developed bifunctional RNAs that target this sequence. Consequently, there is a 2-fold mechanism of SMN induction: inhibition of the intronic repressor and recruitment of SR proteins via the SR recruitment sequence of the bifunctional RNA. The bifunctional RNAs effectively increased SMN in human primary SMA fibroblasts. Lead candidates were synthesized as 2'-O-methyl RNAs and were directly injected in the central nervous system of SMA mice. Single-RNA injections were able to illicit a robust induction of SMN protein in the brain and throughout the spinal column of neonatal SMA mice. In a severe model of SMA, mean life span was extended following the delivery of bifunctional RNAs. This technology has direct implications for the development of an SMA therapy, but also lends itself to a multitude of diseases caused by aberrant pre-mRNA splicing.


Asunto(s)
Terapia Genética , Intrones , Atrofia Muscular Espinal/metabolismo , Atrofia Muscular Espinal/terapia , ARN sin Sentido/uso terapéutico , Secuencias Reguladoras de Ácidos Nucleicos , Proteína 2 para la Supervivencia de la Neurona Motora/genética , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Fibroblastos/metabolismo , Humanos , Ratones , Ratones Transgénicos , Atrofia Muscular Espinal/genética , ARN sin Sentido/química , ARN sin Sentido/genética , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Proteína 2 para la Supervivencia de la Neurona Motora/metabolismo
11.
Life Sci Alliance ; 4(1)2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33234679

RESUMEN

Spinal muscular atrophy (SMA) is a motor neuron disease and the leading genetic cause of infant mortality. SMA results from insufficient survival motor neuron (SMN) protein due to alternative splicing. Antisense oligonucleotides, gene therapy and splicing modifiers recently received FDA approval. Although severe SMA transgenic mouse models have been beneficial for testing therapeutic efficacy, models mimicking milder cases that manifest post-infancy have proven challenging to develop. We established a titratable model of mild and moderate SMA using the splicing compound NVS-SM2. Administration for 30 d prevented development of the SMA phenotype in severe SMA mice, which typically show rapid weakness and succumb by postnatal day 11. Furthermore, administration at day eight resulted in phenotypic recovery. Remarkably, acute dosing limited to the first 3 d of life significantly enhanced survival in two severe SMA mice models, easing the burden on neonates and demonstrating the compound as suitable for evaluation of follow-on therapies without potential drug-drug interactions. This pharmacologically tunable SMA model represents a useful tool to investigate cellular and molecular pathogenesis at different stages of disease.


Asunto(s)
Atrofia Muscular Espinal/tratamiento farmacológico , Atrofia Muscular Espinal/genética , Piperidinas/administración & dosificación , Pirazoles/administración & dosificación , Piridazinas/administración & dosificación , Empalme del ARN/efectos de los fármacos , Animales , Animales Recién Nacidos , Supervivencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Estimación de Kaplan-Meier , Ratones , Ratones Transgénicos , Neuronas Motoras/metabolismo , Atrofia Muscular Espinal/metabolismo , Fenotipo , Proteína 2 para la Supervivencia de la Neurona Motora/genética , Proteína 2 para la Supervivencia de la Neurona Motora/metabolismo , Tiempo de Tratamiento
12.
JCI Insight ; 5(12)2020 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-32516136

RESUMEN

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.


Asunto(s)
Neuronas Motoras/metabolismo , Atrofia Muscular Espinal/genética , ARN Nuclear Pequeño/genética , Sinapsis/metabolismo , Animales , Modelos Animales de Enfermedad , Ratones , Atrofia Muscular Espinal/patología , Empalme del ARN/genética , ARN Mensajero/metabolismo , ARN Nuclear Pequeño/metabolismo , Médula Espinal/metabolismo
13.
Sci Rep ; 9(1): 9472, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-31263170

RESUMEN

Spinal Muscular Atrophy (SMA) is a monogenic neurodegenerative disorder and the leading genetic cause of infantile mortality. While several functions have been ascribed to the SMN (survival motor neuron) protein, their specific contribution to the disease has yet to be fully elucidated. We hypothesized that some, but not all, SMN homologues would rescue the SMA phenotype in mouse models, thereby identifying disease-relevant domains. Using AAV9 to deliver Smn homologs to SMA mice, we identified a conservation threshold that marks the boundary at which homologs can rescue the SMA phenotype. Smn from Danio rerio and Xenopus laevis significantly prevent disease, whereas Smn from Drosophila melanogaster, Caenorhabditis elegans, and Schizosaccharomyces pombe was significantly less efficacious. This phenotypic rescue correlated with correction of RNA processing defects induced by SMN deficiency and neuromuscular junction pathology. Based upon the sequence conservation in the rescuing homologs, a minimal SMN construct was designed consisting of exons 2, 3, and 6, which showed a partial rescue of the SMA phenotype. While a significant extension in survival was observed, the absence of a complete rescue suggests that while the core conserved region is essential, additional sequences contribute to the overall ability of the SMN protein to rescue disease pathology.


Asunto(s)
Atrofia Muscular Espinal/metabolismo , Proteína 1 para la Supervivencia de la Neurona Motora/metabolismo , Animales , Caenorhabditis elegans , Modelos Animales de Enfermedad , Drosophila melanogaster , Evolución Molecular , Ratones , Ratones Noqueados , Atrofia Muscular Espinal/genética , Schizosaccharomyces , Proteína 1 para la Supervivencia de la Neurona Motora/genética , Proteínas de Xenopus/genética , Proteínas de Xenopus/metabolismo , Xenopus laevis , Pez Cebra/genética , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
14.
J Neuromuscul Dis ; 4(3): 237-249, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28598854

RESUMEN

BACKGROUND: Spinal muscular atrophy (SMA) is a neurodegenerative autosomal recessive disorder characterized by the loss of α-motor neurons. A variety of molecular pathways are being investigated to elevate SMN protein expression in SMA models and in the clinic. One of these approaches involves stabilizing the SMNΔ7 protein by inducing translational read-through. Previous studies have demonstrated that functionality and stability are partially restored to the otherwise unstable SMNΔ7 by the addition of non-specific C-terminal peptide sequences, or by inducing a similar molecular event through the use of read-through inducing compounds such as aminoglycosides. OBJECTIVE: The objective was to determine the efficacy of the macrolide Azithromycin (AZM), an FDA approved read-through-inducing compound, in the well-established severe mouse model of SMA. METHODS: Initially, dosing regimen following ICV administrations of AZM at different post-natal days and concentrations was determined by their impact on SMN levels in disease-relevant tissues. Selected dose was then tested for phenotypic parameters changes as compared to the appropriate controls and in conjugation to another therapy. RESULTS: AZM increases SMN protein in disease relevant tissues, however, this did not translate into similar improvements in the SMA phenotype in a severe mouse model of SMA. Co-administration of AZM and a previously developed antisense oligonucleotide that increases SMN2 splicing, resulted in an improvement in the SMA phenotype beyond either AZM or ASO alone, including a highly significant extension in survival with improvement in body weight and movement. CONCLUSIONS: It is important to explore various approaches for SMA therapeutics, hence compounds that specifically induce SMNΔ7 read-through, without having prohibitive toxicity, may provide an alternative platform for a combinatorial treatment. Here we established that AZM activity at a low dose can increase SMN protein in disease-relevant animal model and can impact disease severity.


Asunto(s)
Azitromicina/farmacología , Encéfalo/efectos de los fármacos , Atrofia Muscular Espinal/patología , Fármacos Neuroprotectores/farmacología , Animales , Encéfalo/patología , Modelos Animales de Enfermedad , Ratones , Ratones Transgénicos , Oligonucleótidos Antisentido/farmacología , Médula Espinal/efectos de los fármacos , Médula Espinal/patología
15.
JCI Insight ; 2(5): e89970, 2017 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-28289706

RESUMEN

Spinal muscular atrophy (SMA) is a leading genetic cause of infantile death and is caused by the loss of survival motor neuron-1 (SMN1). Importantly, a nearly identical gene is present called SMN2; however, the majority of SMN2-derived transcripts are alternatively spliced and encode a truncated, dysfunctional protein. Recently, several compounds designed to increase SMN protein have entered clinical trials, including antisense oligonucleotides (ASOs), traditional small molecules, and gene therapy. Expanding beyond SMN-centric therapeutics is important, as it is likely that the breadth of the patient spectrum and the inherent complexity of the disease will be difficult to address with a single therapeutic strategy. Several SMN-independent pathways that could impinge upon the SMA phenotype have been examined with varied success. To identify disease-modifying pathways that could serve as stand-alone therapeutic targets or could be used in combination with an SMN-inducing compound, we investigated adeno-associated virus-mediated (AAV-mediated) gene therapy using plastin-3 (PLS3). Here, we report that AAV9-PLS3 extends survival in an intermediate model of SMA mice as well as in a pharmacologically induced model of SMA using a splice-switching ASO that increases SMN production. PLS3 coadministration improves the phenotype beyond the ASO, demonstrating the potential utility of combinatorial therapeutics in SMA that target SMN-independent and SMN-dependent pathways.


Asunto(s)
Glicoproteínas de Membrana/fisiología , Proteínas de Microfilamentos/fisiología , Atrofia Muscular Espinal/patología , Animales , Dependovirus/genética , Modelos Animales de Enfermedad , Vectores Genéticos , Humanos , Glicoproteínas de Membrana/genética , Ratones , Ratones Noqueados , Proteínas de Microfilamentos/genética , Neuronas Motoras/fisiología , Fibras Musculares Esqueléticas/patología , Análisis de Supervivencia , Proteína 1 para la Supervivencia de la Neurona Motora/genética
16.
EMBO Mol Med ; 5(7): 1103-18, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23740718

RESUMEN

Spinal muscular atrophy (SMA) is a neurodegenerative disease that causes progressive muscle weakness, which primarily targets proximal muscles. About 95% of SMA cases are caused by the loss of both copies of the SMN1 gene. SMN2 is a nearly identical copy of SMN1, which expresses much less functional SMN protein. SMN2 is unable to fully compensate for the loss of SMN1 in motor neurons but does provide an excellent target for therapeutic intervention. Increased expression of functional full-length SMN protein from the endogenous SMN2 gene should lessen disease severity. We have developed and implemented a new high-throughput screening assay to identify small molecules that increase the expression of full-length SMN from a SMN2 reporter gene. Here, we characterize two novel compounds that increased SMN protein levels in both reporter cells and SMA fibroblasts and show that one increases lifespan, motor function, and SMN protein levels in a severe mouse model of SMA.


Asunto(s)
Descubrimiento de Drogas , Atrofia Muscular Espinal/tratamiento farmacológico , Bibliotecas de Moléculas Pequeñas/uso terapéutico , Proteína 2 para la Supervivencia de la Neurona Motora/genética , Regulación hacia Arriba/efectos de los fármacos , Animales , Células Cultivadas , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibroblastos/patología , Ensayos Analíticos de Alto Rendimiento , Humanos , Ratones , Atrofia Muscular Espinal/genética , Atrofia Muscular Espinal/fisiopatología , ARN Mensajero/genética , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/farmacología , Proteína 1 para la Supervivencia de la Neurona Motora/análisis , Proteína 1 para la Supervivencia de la Neurona Motora/genética , Proteína 2 para la Supervivencia de la Neurona Motora/análisis
17.
Hum Gene Ther ; 23(3): 330-5, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22029744

RESUMEN

Spinal muscular atrophy (SMA), an autosomal recessive neuromuscular disorder, is the leading genetic cause of infant mortality. SMA is caused by the homozygous loss of Survival Motor Neuron-1 (SMN1). In humans, a nearly identical copy gene is present, SMN2. SMN2 is retained in all SMA patients and encodes the same protein as SMN1. However, SMN1 and SMN2 differ by a silent C-to-T transition at the 5' end of exon 7, causing alternative splicing of SMN2 transcripts and low levels of full-length SMN. SMA is monogenic and therefore well suited for gene-replacement strategies. Recently, self-complementary adeno-associated virus (scAAV) vectors have been used to deliver the SMN cDNA to an animal model of disease, the SMNΔ7 mouse. In this study, we examine a severe model of SMA, Smn(-/-);SMN2(+/+), to determine whether gene replacement is viable in a model in which disease development begins in utero. Using two delivery paradigms, intracerebroventricular injections and intravenous injections, we delivered scAAV9-SMN and demonstrated a two to four fold increase in survival, in addition to improving many of the phenotypic parameters of the model. This represents the longest extension in survival for this severe model for any therapeutic intervention and suggests that postsymptomatic treatment of SMA may lead to significant improvement of disease severity.


Asunto(s)
Dependovirus/genética , Atrofia Muscular Espinal/terapia , Proteínas del Complejo SMN/genética , Animales , Modelos Animales de Enfermedad , Terapia Genética , Vectores Genéticos , Ratones , Ratones Transgénicos , Atrofia Muscular Espinal/genética , Atrofia Muscular Espinal/metabolismo , Proteínas del Complejo SMN/metabolismo , Índice de Severidad de la Enfermedad
18.
J Vis Exp ; (56)2011 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-21988897

RESUMEN

Despite the protective role that blood brain barrier plays in shielding the brain, it limits the access to the central nervous system (CNS) which most often results in failure of potential therapeutics designed for neurodegenerative disorders. Neurodegenerative diseases such as Spinal Muscular Atrophy (SMA), in which the lower motor neurons are affected, can benefit greatly from introducing the therapeutic agents into the CNS. The purpose of this video is to demonstrate two different injection paradigms to deliver therapeutic materials into neonatal mice soon after birth. One of these methods is injecting directly into cerebral lateral ventricles (Intracerebroventricular) which results in delivery of materials into the CNS through the cerebrospinal fluid. The second method is a temporal vein injection (intravenous) that can introduce different therapeutics into the circulatory system, leading to systemic delivery including the CNS. Widespread transduction of the CNS is achievable if an appropriate viral vector and viral serotype is utilized. Visualization and utilization of the temporal vein for injection is feasible up to postnatal day 6. However, if the delivered material is intended to reach the CNS, these injections should take place while the blood brain barrier is more permeable due to its immature status, preferably prior to postnatal day 2. The fully developed blood brain barrier greatly limits the effectiveness of intravenous delivery. Both delivery systems are simple and effective once the surgical aptitude is achieved. They do not require any extensive surgical devices and can be performed by a single person. However, these techniques are not without challenges. The small size of postnatal day 2 pups and the subsequent small target areas can make the injections difficult to perform and initially challenging to replicate.


Asunto(s)
Inyecciones Intravenosas/métodos , Inyecciones Intravenosas/veterinaria , Inyecciones Intraventriculares/métodos , Inyecciones Intraventriculares/veterinaria , Preparaciones Farmacéuticas/administración & dosificación , Animales , Animales Recién Nacidos , Barrera Hematoencefálica/metabolismo , Inyecciones Intravenosas/instrumentación , Inyecciones Intraventriculares/instrumentación , Ratones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA