Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
J Neurochem ; 153(2): 264-275, 2020 04.
Article in English | MEDLINE | ID: mdl-31811660

ABSTRACT

Spinal muscular atrophy (SMA) is a motor neuron disease caused by loss of function mutations in the Survival Motor Neuron 1 (SMN1) gene and reduced expression of the SMN protein, leading to spinal motor neuron death, muscle weakness and atrophy. Although humans harbour the highly homologous SMN2 gene, its defective splicing regulation yields a truncated and unstable SMN protein. The first therapy for SMA was recently approved by the Food and Drug Administration and consists of an antisense oligonucleotide (Nusinersen) rendering SMN2 functional and thus improving patients' motor activity and quality of life. Nevertheless, not all patients equally respond to this therapy and the long-term tolerability and safety of Nusinersen are still unknown. Herein, in vivo splicing assays indicated that the HDAC inhibitor LBH589 is particularly efficient in rescuing the SMN2 splicing defect in SMA fibroblasts and SMA type-I mice-derived neural stem cells. Western blot analyses showed that LBH589 also causes a significant increase in SMN protein expression in SMA cells. Moreover chromatin immunoprecipitation analyses revealed that LBH589 treatment induces widespread H4 acetylation of the entire SMN2 locus and selectively favors the inclusion of the disease-linked exon 7 in SMN2 mature mRNA. The combined treatment of SMA cells with sub-optimal doses of LBH589 and of an antisense oligonucleotide that mimic Nusinersen (ASO_ISSN1) elicits additive effects on SMN2 splicing and SMN protein expression. These findings suggest that HDAC inhibitors can potentiate the activity of Nusinersen and support the notion that 'SMN-plus' combinatorial therapeutic approaches might represent an enhanced opportunity in the scenario of SMA therapy.


Subject(s)
Muscular Atrophy, Spinal , Oligonucleotides/pharmacology , Panobinostat/pharmacology , RNA Splicing/drug effects , Survival of Motor Neuron 2 Protein/biosynthesis , Animals , Drug Therapy, Combination , Female , Fibroblasts/drug effects , Histone Deacetylase Inhibitors/pharmacology , Humans , Male , Mice , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/metabolism , Neural Stem Cells/drug effects , Oligonucleotides, Antisense/pharmacology , RNA Splicing/genetics , Survival of Motor Neuron 2 Protein/genetics
2.
Stem Cells Dev ; 28(7): 438-453, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30667343

ABSTRACT

Spinal muscular atrophy (SMA) is caused by the mutation or deletion of the survival motor neuron 1 (SMN1) gene. Only ∼10% of the products of SMN2, a paralogue of SMN1, are functional full-length SMN (SMN-FL) proteins, whereas SMN2 primarily produces alternatively spliced transcripts lacking exon 7. Reduced SMN protein levels in SMA patients lead to progressive degeneration of spinal motor neurons (MNs). In this study, we report an advanced platform based on an SMN2 splicing-targeting approach for SMA drug screening and validation using an SMN2 splicing reporter cell line and an in vitro human SMA model through induced pluripotent stem cell (iPSC) technology. Through drug screening using a robust cell-based luciferase assay to quantitatively measure SMN2 splicing, the small-molecule candidate compound rigosertib was identified as an SMN2 splicing modulator that led to enhanced SMN protein expression. The therapeutic potential of the candidate compound was validated in MN progenitors differentiated from SMA patient-derived iPSCs (SMA iPSC-pMNs) as an in vitro human SMA model, which recapitulated the biochemical and molecular phenotypes of SMA, including lower levels of SMN-FL transcripts and protein, enhanced cell death, and reduced neurite length. The candidate compound exerted strong splicing correction activity for SMN2 and potently alleviated the disease-related phenotypes of SMA iPSC-pMNs by modulating various cellular and molecular abnormalities. Our combined screening platform representing a pMN model of human SMA provides an efficient and reliable drug screening system and is a promising resource for drug evaluation and the exploration of drug modes of action.


Subject(s)
Alternative Splicing/drug effects , Glycine/analogs & derivatives , Models, Neurological , Muscular Atrophy, Spinal , Sulfones/pharmacology , Animals , Cell Line , Glycine/pharmacology , Humans , Mice , Mice, Transgenic , Muscular Atrophy, Spinal/drug therapy , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/metabolism , Muscular Atrophy, Spinal/pathology , Survival of Motor Neuron 2 Protein/biosynthesis , Survival of Motor Neuron 2 Protein/genetics
3.
Drugs Today (Barc) ; 53(6): 327-337, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28799578

ABSTRACT

Patients with spinal muscular atrophy (SMA) have an autosomal recessive disease that limits their ability to produce survival motor neuron (SMN) protein in the CNS resulting in progressive wasting of voluntary muscles. Detailed studies over several years have demonstrated that phosphorothioate and 2'-O-methoxyethyl- modified antisense oligonucleotides (ASOs) targeting the ISS-N1 site increase SMN2 exon 7 inclusion, thus increasing levels of SMN protein in a dose- and time-dependent manner in liver, kidney and skeletal muscle, and CNS tissues only when administered intrathecally. On a dose basis, nusinersen was found to be the most potent ASO for SMN2 splicing correction in the CNS of adult mice. After nusinersen was found to increase levels of SMN protein in the CNS of mice and subhuman primates without causing significant adverse events, it was advanced into clinical studies in patients with SMA. These trials in SMA patients have demonstrated significant improvements in various measures of motor function and in progression to movement developments not normally seen in SMA patients. In addition, there have been significant extensions in life expectancy. These findings led to the U.S. and European approval of nusinersen for use in SMA patients of all ages.


Subject(s)
Alternative Splicing/drug effects , Oligonucleotides, Antisense/therapeutic use , Oligonucleotides/therapeutic use , Spinal Muscular Atrophies of Childhood/therapy , Adult , Alternative Splicing/genetics , Animals , Central Nervous System/metabolism , Clinical Trials as Topic , Drug Evaluation, Preclinical , Exons , Gene Dosage , Haplorhini , Humans , Infant , Injections, Spinal , Kidney Diseases/chemically induced , Mice , Multicenter Studies as Topic , Oligonucleotides/administration & dosage , Oligonucleotides/adverse effects , Oligonucleotides/pharmacokinetics , Oligonucleotides, Antisense/administration & dosage , Oligonucleotides, Antisense/adverse effects , Oligonucleotides, Antisense/pharmacokinetics , Protein Stability , Spinal Muscular Atrophies of Childhood/genetics , Survival of Motor Neuron 1 Protein/genetics , Survival of Motor Neuron 2 Protein/biosynthesis , Survival of Motor Neuron 2 Protein/genetics , Thionucleotides/administration & dosage , Thionucleotides/adverse effects , Thionucleotides/pharmacokinetics , Thionucleotides/therapeutic use , Thrombocytopenia/chemically induced , Up-Regulation/drug effects
4.
Rev Neurol ; 64(s03): S19-S24, 2017 May 17.
Article in Spanish | MEDLINE | ID: mdl-28524214

ABSTRACT

The new treatments of spinal muscular atrophy (SMA) due by SMN1 gene deletions are reviewed. There are several ways to increase the protein SMN, its activity and persistence in the tissues. Neuroprotective drugs as olesoxime or riluzole, and drugs acting by epigenetic mechanisms, as histone deacetylase inhibitors, have shown positive effects in preclinical studies but no clear efficacy in clinical trials. They might give in the future added benefits when used associated to other genetic modifying drugs. The best improvements in murine models of SMA and in clinical trials have been reached with antisense oligonucleotides, drugs that modify the splicing of SMN2, and they are expected to get better in the near future. Nusinersen, a methoxi-ethyl phosphotioate antisense oligonucleotide has recently approved for treatment of patients with SMA type 1 after having proved its efficacy in clinical trial phase 3. The results of nusinersen are reviewed. New modifications of antisense oligonucleotides with better access to brain, spinal cord and peripheral tissues are on the way. There are data of the efficacy of the genetic therapy with SMN1 gene through adenoassociated virus, now in phase 1 trial. A constant feature of these new treatments is that the earlier the treatment, the best are the results, and they are even better in presymptomatic stage. The general standards of care, particularly nutrition and respiratory management are needed in order to reach optimal results with the new therapies.


TITLE: Posibilidades de tratamiento en la atrofia espinal infantil.Se revisan los nuevos tratamientos de la atrofia muscular espinal (AME) producida por delecion del gen SMN1. Se describen las diferentes posibilidades de incrementar la proteina SMN, de su actividad y persistencia en el organismo. Farmacos neuroprotectores, como olesoxime y riluzol, y farmacos que actuan epigeneticamente, como inhibidores de histona deacetilasa, han mostrado cierto efecto positivo en fases preclinicas pero no han conseguido eficacia en los ensayos clinicos. Podrian proporcionar en un futuro un beneficio añadidos a otros farmacos modificadores geneticos. Los mayores cambios en estudios de modelos del raton SMA y en fases clinicas se han encontrado con oligonucleotidos antisentido que modifican el splicing del gen SMN2, y se espera que mejoren en el futuro proximo. Recientemente se ha aprobado el nusinersen, un metoxietilo fosforotioato-oligonucleotido antisentido, para uso en pacientes con AME de tipo I una vez demostrada su eficacia en pacientes en el ensayo en fase 3. Se revisan los resultados de este farmaco. Estan en marcha modificaciones de oligonucleotidos antisentido que amplien la liberacion en el sistema nervioso y en tejidos perifericos. Hay datos que sugieren eficacia de la terapia genica introduciendo el gen SMN1 mediante virus adenoasociados, actualmente en fase clinica 1. Una constante en estos nuevos tratamientos es que los resultados se optimizan en las etapas precoces de la enfermedad y, mejor aun, en estadio presintomatico. Se subraya la importancia de los cuidados generales optimos, especialmente nutricionales y respiratorios, para conseguir los mejores resultados con las nuevas terapias.


Subject(s)
Spinal Muscular Atrophies of Childhood/therapy , Therapies, Investigational , Animals , Child , Clinical Trials as Topic , Dependovirus/genetics , Disease Models, Animal , Epigenesis, Genetic , Gene Deletion , Genetic Therapy , Genetic Vectors/therapeutic use , Histone Deacetylase Inhibitors/therapeutic use , Humans , Mice , Mice, Neurologic Mutants , Multicenter Studies as Topic , Neuroprotective Agents/therapeutic use , Oligonucleotides/therapeutic use , Oligonucleotides, Antisense/therapeutic use , Palliative Care , Pluripotent Stem Cells/transplantation , RNA Splicing , Recombinant Proteins/genetics , Spinal Muscular Atrophies of Childhood/genetics , Survival of Motor Neuron 1 Protein/biosynthesis , Survival of Motor Neuron 1 Protein/genetics , Survival of Motor Neuron 2 Protein/biosynthesis , Survival of Motor Neuron 2 Protein/genetics
6.
PLoS One ; 10(3): e0120721, 2015.
Article in English | MEDLINE | ID: mdl-25781985

ABSTRACT

Spinal muscular atrophy (SMA) is an inherited neuromuscular disease caused by deletion or mutation of SMN1 gene. All SMA patients carry a nearly identical SMN2 gene, which produces low level of SMN protein due to mRNA exon 7 exclusion. Previously, we found that the testis of SMA mice (smn-/- SMN2) expresses high level of SMN2 full-length mRNA, indicating a testis-specific mechanism for SMN2 exon 7 inclusion. To elucidate the underlying mechanism, we established primary cultures of testis cells from SMA mice and analyzed them for SMN2 exon 7 splicing. We found that primary testis cells after a 2-hour culture still expressed high level of SMN2 full-length mRNA, but the level decreased after longer cultures. We then compared the protein levels of relevant splicing factors, and found that the level of Tra2-ß1 also decreased during testis cell culture, correlated with SMN2 full-length mRNA downregulation. In addition, the testis of SMA mice expressed the highest level of Tra2-ß1 among the many tissues examined. Furthermore, overexpression of Tra2-ß1, but not ASF/SF2, increased SMN2 minigene exon 7 inclusion in primary testis cells and spinal cord neurons, whereas knockdown of Tra2-ß1 decreased SMN2 exon 7 inclusion in primary testis cells of SMA mice. Therefore, our results indicate that high expression level of Tra2-ß1 is responsible for increased SMN2 exon 7 inclusion in the testis of SMA mice. This study also suggests that the expression level of Tra2-ß1 may be a modifying factor of SMA disease and a potential target for SMA treatment.


Subject(s)
Exons , Muscular Atrophy, Spinal/metabolism , Nuclear Proteins/metabolism , RNA Splicing , RNA-Binding Proteins/metabolism , Survival of Motor Neuron 2 Protein/biosynthesis , Testis/metabolism , Animals , Cells, Cultured , Male , Mice , Mice, Knockout , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/pathology , Nuclear Proteins/genetics , RNA-Binding Proteins/genetics , Serine-Arginine Splicing Factors , Survival of Motor Neuron 2 Protein/genetics
7.
Br J Pharmacol ; 166(3): 1114-26, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22220673

ABSTRACT

BACKGROUND AND PURPOSE: Spinal muscular atrophy (SMA) is a progressive neuromuscular disease. Since disease severity is related to the amount of survival motor neuron (SMN) protein, up-regulated functional SMN protein levels from the SMN2 gene are considered a major SMA drug-discovery strategy. In this study, we investigated the possible effects of triptolide, a diterpene triepoxide purified from Tripterygium wilfordii Hook. F., as a new compound for increasing SMN protein. EXPERIMENTAL APPROACH: The effects and mechanisms of triptolide on the production of SMA protein were determined by cell-based assays using the motor neuronal cell line NSC34 and skin fibroblasts from SMA patients. Wild-type (Smn(+/+) SMN2(-/-) , C57BL/6) and SMA-like (Smn(-/-) SMN2) mice were injected with triptolide (0.01 or 0.1 mg·kg(-1) ·day(-1) , i.p.) and their survival rate and level of change in SMN protein in neurons and muscle tissue measured. KEY RESULTS: In NSC34 cells and human SMA fibroblasts, pM concentrations of triptolide significantly increased SMN protein expression and the levels of SMN complex component (Gemin2 and Gemin3). In human SMA fibroblasts, triptolide increased SMN-containing nuclear gems and the ratio of full-length transcripts (FL-SMN2) to SMN2 transcripts lacking exon 7 (SMN2Δ7). Furthermore, in SMA-like mice, triptolide significantly increased SMN protein levels in the brain, spinal cord and gastrocnemius muscle. Furthermore, triptolide treatment increased survival and reduced weight loss in SMA-like mice. CONCLUSION AND IMPLICATIONS: Triptolide enhanced SMN protein production by promoting SMN2 activation, exon 7 inclusion and increasing nuclear gems, and increased survival in SMA mice, which suggests triptolide might be a potential candidate for SMA therapy.


Subject(s)
Diterpenes/therapeutic use , Fibroblasts/drug effects , Motor Neurons/drug effects , Muscular Atrophy, Spinal/drug therapy , Neuroprotective Agents/therapeutic use , Phenanthrenes/therapeutic use , Survival of Motor Neuron 2 Protein/biosynthesis , Transcription, Genetic/drug effects , Animals , Apoptosis/drug effects , Blotting, Western , Body Weight/drug effects , Cell Culture Techniques , Cell Line , Cell Survival/drug effects , Disease Models, Animal , Diterpenes/administration & dosage , Diterpenes/isolation & purification , Diterpenes/pharmacology , Dose-Response Relationship, Drug , Epoxy Compounds/administration & dosage , Epoxy Compounds/isolation & purification , Epoxy Compounds/pharmacology , Epoxy Compounds/therapeutic use , Fibroblasts/metabolism , Fibroblasts/pathology , Gemini of Coiled Bodies/drug effects , Gemini of Coiled Bodies/metabolism , Humans , Kaplan-Meier Estimate , Mice , Mice, Knockout , Molecular Structure , Motor Neurons/metabolism , Muscular Atrophy, Spinal/metabolism , Muscular Atrophy, Spinal/pathology , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/isolation & purification , Neuroprotective Agents/pharmacology , Phenanthrenes/administration & dosage , Phenanthrenes/isolation & purification , Phenanthrenes/pharmacology , Real-Time Polymerase Chain Reaction , Survival of Motor Neuron 2 Protein/genetics , Tripterygium/chemistry , Up-Regulation
8.
Brain Dev ; 34(3): 213-22, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21561730

ABSTRACT

Spinal muscular atrophy (SMA) is a common autosomal recessive neuromuscular disorder that is caused by loss of the survival motor neuron gene, SMN1. SMA treatment strategies have focused on production of the SMN protein from the almost identical gene, SMN2. Valproic acid (VPA) is a histone deacetylase inhibitor that can increase SMN levels in some SMA cells or SMA patients through activation of SMN2 transcription or splicing correction of SMN2 exon 7. It remains to be clarified what concentration of VPA is required and by what mechanisms the SMN production from SMN2 is elicited. We observed that in two fibroblast cell lines from Japanese SMA patients, more than 1mM of VPA increased SMN2 expression at both the transcript and protein levels. VPA increased not only full-length (FL) transcript level but also exon 7-excluding (Δ7) transcript level in the cell lines and did not change the ratio of FL/Δ7, suggesting that SMN2 transcription was mainly activated. We also found that VPA modulated splicing factor expression: VPA increased the expression of splicing factor 2/alternative splicing factor (SF2/ASF) and decreased the expression of heterogeneous nuclear ribonucleoprotein A1 (hnRNPA1). In conclusion, more than 1mM of VPA activated SMN2 transcription and modulated the expression of splicing factors in our SMA fibroblast cell lines.


Subject(s)
Gene Expression/drug effects , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/biosynthesis , Neuroprotective Agents/pharmacology , Nuclear Proteins/biosynthesis , RNA-Binding Proteins/biosynthesis , Valproic Acid/pharmacology , Adult , Blotting, Western , Cell Line , Fibroblasts/drug effects , Fibroblasts/metabolism , Heterogeneous Nuclear Ribonucleoprotein A1 , Humans , Infant , Muscular Atrophy, Spinal/metabolism , Real-Time Polymerase Chain Reaction , Serine-Arginine Splicing Factors , Survival of Motor Neuron 2 Protein/biosynthesis , Transcription, Genetic/drug effects
9.
RNA Biol ; 8(4): 600-6, 2011.
Article in English | MEDLINE | ID: mdl-21654213

ABSTRACT

Humans have two nearly identical copies of the Survival Motor Neuron (SMN) gene: SMN1 and SMN2. The two SMN genes code for identical proteins; however, SMN2 predominantly generates a shorter transcript due to skipping of exon 7, the last coding exon. Skipping of SMN2 exon 7 leads to production of a truncated SMN protein that is highly unstable. The inability of SMN2 to compensate for the loss of SMN1 results in spinal muscular atrophy (SMA), the second most prevalent genetic cause of infant mortality. Since SMN2 is almost universally present in SMA patients, correction of SMN2 exon 7 splicing holds the promise for cure. Consistently, SMN2 exon 7 splicing has emerged as one of the best studied splicing systems in humans. The vast amount of recent literature provides a clue that SMN2 exon 7 splicing is regulated by an intron definition mechanism, which does not require cross-exon communication as prerequisite for exon inclusion. Our conclusion is based on the prominent role of intronic cis-elements, some of them have emerged as the frontrunners among potential therapeutic targets of SMA. Further, the widely expressed T-cell-restricted intracellular antigen-1 (TIA1), a member of the Q-rich domain containing RNA-binding proteins, has recently been found to regulate SMN exon 7 splicing by binding to intron 7 sequences away from the 5' ss. These findings make a strong argument for an "intron definition model", according to which regulatory sequences within a downstream intron are capable of enforcing exon inclusion even in the absence of a defined upstream 3' ss of an alternatively spliced exon.


Subject(s)
Alternative Splicing , Introns , Muscular Atrophy, Spinal/genetics , Survival of Motor Neuron 1 Protein/genetics , Exons , Humans , Models, Genetic , Poly(A)-Binding Proteins/genetics , Poly(A)-Binding Proteins/metabolism , Survival of Motor Neuron 1 Protein/biosynthesis , Survival of Motor Neuron 1 Protein/metabolism , Survival of Motor Neuron 2 Protein/biosynthesis , Survival of Motor Neuron 2 Protein/genetics , Survival of Motor Neuron 2 Protein/metabolism , T-Cell Intracellular Antigen-1
10.
J Neurosci ; 30(34): 11288-99, 2010 Aug 25.
Article in English | MEDLINE | ID: mdl-20739549

ABSTRACT

Spinal muscular atrophy (SMA), a lethal neurodegenerative disease that occurs in childhood, is caused by the misexpression of the survival of motor neuron (SMN) protein in motor neurons. It is still unclear whether activating motor units in SMA corrects the delay in the postnatal maturation of the motor unit resulting in an enhanced neuroprotection. In the present work, we demonstrate that an adequate NMDA receptor activation in a type 2 SMA mouse model significantly accelerated motor unit postnatal maturation, counteracted apoptosis in the spinal cord, and induced a marked increase of SMN expression resulting from a modification of SMN2 gene transcription pattern. These beneficial effects were dependent on the level of NMDA receptor activation since a treatment with high doses of NMDA led to an acceleration of the motor unit maturation but favored the apoptotic process and decreased SMN expression. In addition, these results suggest that the NMDA-induced acceleration of motor unit postnatal maturation occurred independently of SMN. The NMDA receptor activating treatment strongly extended the life span in two different mouse models of severe SMA. The analysis of the intracellular signaling cascade that lay downstream the activated NMDA receptor revealed an unexpected reactivation of the CaMKII/AKT/CREB (cAMP response element-binding protein) pathway that induced an enhanced SMN expression. Therefore, pharmacological activation of spinal NMDA receptors could constitute a useful strategy for both increasing SMN expression and limiting motor neuron death in SMA spinal cord.


Subject(s)
Motor Neurons/physiology , Muscle Fibers, Skeletal/physiology , Muscular Atrophy, Spinal/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Spinal Cord/growth & development , Survival of Motor Neuron 2 Protein/biosynthesis , Animals , Coculture Techniques , Female , Gene Expression Regulation , Humans , Male , Mice , Mice, Knockout , Mice, Transgenic , Motor Neurons/drug effects , Muscle Fibers, Skeletal/drug effects , Muscular Atrophy, Spinal/pathology , Muscular Atrophy, Spinal/prevention & control , N-Methylaspartate/pharmacology , N-Methylaspartate/therapeutic use , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Receptors, N-Methyl-D-Aspartate/agonists , Severity of Illness Index , Spinal Cord/drug effects
11.
J Neurol Sci ; 287(1-2): 27-31, 2009 Dec 15.
Article in English | MEDLINE | ID: mdl-19758605

ABSTRACT

Childhood spinal muscular atrophy (SMA) is an autosomal recessive disorder characterised by loss of the alpha motor neurones of the spinal cord. SMA is cause by mutations in the survival motor neuron (SMN) gene. There are two copies of the SMN gene: SMN1 and SMN2. The two genes differ by only 11 nucleotides at the genomic level. One of these is a C to T single nucleotide polymorphism (SNP) at position 6 in exon 7. This change alters an exon splicing enhancer in exon 7, meaning that while SMN1 expresses exclusively full-length protein containing exon 7, SMN2 is predominantly alternatively spliced and expresses a truncated transcript lacking exon 7 (SMN7). As all SMA patients are effectively null for SMN1 but retain at least one copy of SMN2, patients express considerably lower levels of functional SMN protein compared with uneffected individuals. Therefore, SMA is triggered by a fall in the levels of expressed full-length protein, and the levels expressed by the retained SMN2 gene control the severity. As a result, RNA manipulation to suppress the alternative splicing event and thus increase SMN exon 7 inclusion has emerged as an attractive therapeutic approach. In this review we have discussed the current state of bifunctional RNAs as a viable therapy, concentrating on recent advances and overall implications of this research on SMA.


Subject(s)
Genetic Therapy/methods , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/therapy , RNA/genetics , Survival of Motor Neuron 1 Protein/genetics , Survival of Motor Neuron 2 Protein/genetics , Alternative Splicing/genetics , Animals , Exons/genetics , Gene Expression Regulation/genetics , Humans , Muscular Atrophy, Spinal/physiopathology , Polymorphism, Single Nucleotide/genetics , Survival of Motor Neuron 1 Protein/biosynthesis , Survival of Motor Neuron 2 Protein/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...