Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Genes Dev ; 34(11-12): 785-805, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32381627

ABSTRACT

Dysregulation of the DNA/RNA-binding protein FUS causes certain subtypes of ALS/FTD by largely unknown mechanisms. Recent evidence has shown that FUS toxic gain of function due either to mutations or to increased expression can disrupt critical cellular processes, including mitochondrial functions. Here, we demonstrate that in human cells overexpressing wild-type FUS or expressing mutant derivatives, the protein associates with multiple mRNAs, and these are enriched in mRNAs encoding mitochondrial respiratory chain components. Notably, this sequestration leads to reduced levels of the encoded proteins, which is sufficient to bring about disorganized mitochondrial networks, reduced aerobic respiration and increased reactive oxygen species. We further show that mutant FUS associates with mitochondria and with mRNAs encoded by the mitochondrial genome. Importantly, similar results were also observed in fibroblasts derived from ALS patients with FUS mutations. Finally, we demonstrate that FUS loss of function does not underlie the observed mitochondrial dysfunction, and also provides a mechanism for the preferential sequestration of the respiratory chain complex mRNAs by FUS that does not involve sequence-specific binding. Together, our data reveal that respiratory chain complex mRNA sequestration underlies the mitochondrial defects characteristic of ALS/FTD and contributes to the FUS toxic gain of function linked to this disease spectrum.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/physiopathology , Gene Expression Regulation/genetics , Mitochondria/pathology , RNA, Messenger/metabolism , RNA-Binding Protein FUS/genetics , RNA-Binding Protein FUS/metabolism , Cell Line , Cell Respiration/genetics , Cells, Cultured , Electron Transport/genetics , Genome, Mitochondrial , Humans , Mitochondria/genetics , Mutation , Protein Aggregation, Pathological/genetics , Protein Binding/genetics
2.
Genes Dev ; 29(16): 1696-706, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-26251528

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is caused by mutations in a number of genes, including the gene encoding the RNA/DNA-binding protein translocated in liposarcoma or fused in sarcoma (TLS/FUS or FUS). Previously, we identified a number of FUS target genes, among them MECP2. To investigate how ALS mutations in FUS might impact target gene expression, we examined the effects of several FUS derivatives harboring ALS mutations, such as R521C (FUS(C)), on MECP2 expression in transfected human U87 cells. Strikingly, FUS(C) and other mutants not only altered MECP2 alternative splicing but also markedly increased mRNA abundance, which we show resulted from sharply elevated stability. Paradoxically, however, MeCP2 protein levels were significantly reduced in cells expressing ALS mutant derivatives. Providing a parsimonious explanation for these results, biochemical fractionation and in vivo localization studies revealed that MECP2 mRNA colocalized with cytoplasmic FUS(C) in insoluble aggregates, which are characteristic of ALS mutant proteins. Together, our results establish that ALS mutations in FUS can strongly impact target gene expression, reflecting a dominant effect of FUS-containing aggregates.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Gene Expression Regulation/genetics , RNA-Binding Protein FUS/genetics , RNA-Binding Protein FUS/metabolism , Amyotrophic Lateral Sclerosis/physiopathology , Cell Line, Tumor , Cytoplasm/metabolism , Humans , Methyl-CpG-Binding Protein 2/genetics , Methyl-CpG-Binding Protein 2/metabolism , Mutation , Protein Transport , RNA Splicing/genetics , RNA Stability/genetics , RNA, Messenger/metabolism
3.
J Vis Exp ; (56)2011 Oct 03.
Article in English | MEDLINE | ID: mdl-21988897

ABSTRACT

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.


Subject(s)
Injections, Intravenous/methods , Injections, Intravenous/veterinary , Injections, Intraventricular/methods , Injections, Intraventricular/veterinary , Pharmaceutical Preparations/administration & dosage , Animals , Animals, Newborn , Blood-Brain Barrier/metabolism , Injections, Intravenous/instrumentation , Injections, Intraventricular/instrumentation , Mice
4.
Wiley Interdiscip Rev RNA ; 2(4): 546-64, 2011.
Article in English | MEDLINE | ID: mdl-21957043

ABSTRACT

Ribonucleoprotein (RNP) complexes function in nearly every facet of cellular activity. The spliceosome is an essential RNP that accurately identifies introns and catalytically removes the intervening sequences, providing exquisite control of spatial, temporal, and developmental gene expressions. U-snRNPs are the building blocks for the spliceosome. A significant amount of insight into the molecular assembly of these essential particles has recently come from a seemingly unexpected area of research: neurodegeneration. Survival motor neuron (SMN) performs an essential role in the maturation of snRNPs, while the homozygous loss of SMN1 results in the development of spinal muscular atrophy (SMA), a devastating neurodegenerative disease. In this review, the function of SMN is examined within the context of snRNP biogenesis and evidence is examined which suggests that the SMN functional defects in snRNP biogenesis may account for the motor neuron pathology observed in SMA.


Subject(s)
Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/metabolism , Ribonucleoproteins, Small Nuclear/biosynthesis , Survival of Motor Neuron 1 Protein/genetics , Survival of Motor Neuron 1 Protein/metabolism , Alternative Splicing , Axons/metabolism , Humans , Models, Neurological , Muscular Atrophy, Spinal/classification , Muscular Atrophy, Spinal/etiology , Protein Structure, Tertiary , RNA Precursors/genetics , RNA Precursors/metabolism , RNA Splicing , Survival of Motor Neuron 1 Protein/chemistry , Survival of Motor Neuron 2 Protein/genetics , Survival of Motor Neuron 2 Protein/metabolism
5.
J Neurosci ; 30(1): 126-30, 2010 Jan 06.
Article in English | MEDLINE | ID: mdl-20053895

ABSTRACT

Spinal muscular atrophy is a leading genetic cause of infantile death and occurs in approximately 1/6000 live births. SMA is caused by the loss of Survival Motor Neuron-1 (SMN1), however, all patients retain at least one copy of a nearly identical gene called SMN2. While SMN2 and SMN1 are comprised of identical coding sequences, the majority of SMN2 transcripts are alternatively spliced, encoding a truncated protein that is unstable and nonfunctional. Considerable effort has focused upon modulating the SMN2 alternative splicing event since this would produce more wild-type protein. Recently we reported the development of an optimized trans-splicing system that involved the coexpression of a SMN2 trans-splicing RNA and an antisense RNA that blocks a downstream splice site in SMN2 pre-mRNA. Here, we demonstrate that in vivo delivery of the optimized trans-splicing vector increases an important SMN-dependent activity, snRNP assembly, in disease-relevant tissue in the SMA mouse model. A single injection of the vector into the intracerebral-ventricular space in SMA neonates also lessens the severity of the SMA phenotype in a severe SMA mouse model, extending survival approximately 70%. Collectively, these results provide the first in vivo demonstration that SMN2 trans-splicing leads to a lessening of the severity of the SMA phenotype and provide evidence for the power of this strategy for reprogramming genetic diseases at the pre-mRNA level.


Subject(s)
Disease Models, Animal , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/therapy , Trans-Splicing/physiology , Animals , Animals, Newborn , Gene Transfer Techniques , Genetic Therapy/methods , Injections, Intraventricular , Mice , Mice, Knockout , Muscular Atrophy, Spinal/pathology , Severity of Illness Index , Survival of Motor Neuron 1 Protein/administration & dosage , Survival of Motor Neuron 1 Protein/genetics , Survival of Motor Neuron 2 Protein/administration & dosage , Survival of Motor Neuron 2 Protein/genetics , Trans-Splicing/genetics
6.
PLoS One ; 3(10): e3468, 2008.
Article in English | MEDLINE | ID: mdl-18941511

ABSTRACT

RNA modalities are developing as a powerful means to re-direct pathogenic pre-mRNA splicing events. Improving the efficiency of these molecules in vivo is critical as they move towards clinical applications. Spinal muscular atrophy (SMA) is caused by loss of SMN1. A nearly identical copy gene called SMN2 produces low levels of functional protein due to alternative splicing. We previously reported a trans-splicing RNA (tsRNA) that re-directed SMN2 splicing. Now we show that reducing the competition between endogenous splices sites enhanced the efficiency of trans-splicing. A single vector system was developed that expressed the SMN tsRNA and a splice-site blocking antisense (ASO-tsRNA). The ASO-tsRNA vector significantly elevated SMN levels in primary SMA patient fibroblasts, within the central nervous system of SMA mice and increased SMN-dependent in vitro snRNP assembly. These results demonstrate that the ASO-tsRNA strategy provides insight into the trans-splicing mechanism and a means of significantly enhancing trans-splicing activity in vivo.


Subject(s)
RNA, Messenger/genetics , Survival of Motor Neuron 2 Protein/genetics , Trans-Splicing , Animals , Cell Line , Cells, Cultured , Central Nervous System , Fibroblasts/pathology , Humans , Mice , Models, Animal , Muscular Atrophy, Spinal/genetics , RNA, Antisense/pharmacology , Transfection
7.
Biochem Biophys Res Commun ; 375(1): 119-23, 2008 Oct 10.
Article in English | MEDLINE | ID: mdl-18680723

ABSTRACT

Spinal muscular atrophy (SMA) is a severe neuromuscular disease characterized by loss of spinal alpha-motor neurons, resulting in the paralysis of skeletal muscle. SMA is caused by deficiency of survival motor neuron (SMN) protein levels. Recent evidence has highlighted an axon-specific role for SMN protein, raising the possibility that axon degeneration may be an early event in SMA pathogenesis. The Wallerian degeneration slow (Wld(s)) gene is a spontaneous dominant mutation in mice that delays axon degeneration by approximately 2-3 weeks. We set out to examine the effect of Wld(s) on the phenotype of a mouse model of SMA. We found that Wld(s) does not alter the SMA phenotype, indicating that Wallerian degeneration does not directly contribute to the pathogenesis of SMA development.


Subject(s)
Genes, Dominant , Muscular Atrophy, Spinal/etiology , Nerve Tissue Proteins/genetics , Wallerian Degeneration/genetics , Animals , Anterior Horn Cells/pathology , Disease Models, Animal , Disease Progression , Mice , Mice, Mutant Strains , Muscular Atrophy, Spinal/pathology , Sciatic Nerve/pathology , Spinal Nerve Roots/pathology , Wallerian Degeneration/complications
8.
Mol Ther ; 15(8): 1471-8, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17551501

ABSTRACT

Spinal muscular atrophy (SMA) is caused by loss of survival motor neuron-1 (SMN1). A nearly identical copy gene called SMN2 is present in all SMA patients; however SMN2 produces low levels of functional protein due to alternative splicing. Recently a therapeutic approach has been developed referred to as trans-splicing. Conceptually, this strategy relies upon pre-messenger RNA (pre-mRNA) splicing occurring between two separate molecules: (i) the endogenous target RNA and (ii) the therapeutic RNA that provides the correct RNA sequence via a trans-splicing event. SMN trans-splicing RNAs were initially examined and expressed from a plasmid-backbone and shown to re-direct splicing from a SMN2 mini-gene as well as from endogenous transcripts. Subsequently, recombinant adeno-associated viral vectors were developed that expressed and delivered trans-splicing RNAs to SMA patient fibroblasts. In the severe SMA patient fibroblasts, SMN2 splicing was redirected via trans-splicing to produce increased levels of full-length SMN mRNA and total SMN protein levels. Finally, small nuclear ribonucleoprotein (snRNP) assembly, a critical function of SMN, was restored to SMN-deficient SMA fibroblasts following treatment with the trans-splicing vector. Together these results demonstrate that the alternatively spliced SMN2 exon 7 is a tractable target for replacement by trans-splicing.


Subject(s)
Alternative Splicing/genetics , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Trans-Splicing/genetics , Base Sequence , Cell Line , Cell Survival , Fibroblasts , Gene Transfer Techniques , Humans , Molecular Sequence Data , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/metabolism , Muscular Atrophy, Spinal/pathology , RNA, Messenger/genetics , SMN Complex Proteins , Survival of Motor Neuron 1 Protein , Survival of Motor Neuron 2 Protein , Transcription, Genetic/genetics
9.
Mol Ther ; 14(1): 54-62, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16580882

ABSTRACT

Spinal muscular atrophy (SMA) is an autosomal recessive neuromuscular disorder that is the leading genetic cause of infant mortality. SMA is caused by the loss of survival motor neuron-1 (SMN1). In humans, a nearly identical copy gene is present, called SMN2. SMN2 is retained in all SMA patients and encodes an identical protein compared to SMN1. However, a single silent nucleotide difference in SMN2 exon 7 results in the production of a spliced isoform (called SMNDelta7) that encodes a nonfunctional protein. The presence of SMN2 represents a unique therapeutic target since SMN2 has the capacity to encode a fully functional protein. Here we describe an in vivo delivery system for short bifunctional RNAs that modulate SMN2 splicing. Bifunctional RNAs derive their name from the presence of two domains: an antisense RNA sequence specific to a target RNA and an untethered RNA segment that serves as a binding platform for splicing factors. Plasmid-based and recombinant adeno-associated virus vectors were developed that expressed bifunctional RNAs that stimulated SMN2 exon 7 inclusion and full-length SMN protein in patient fibroblasts. These experiments provide a mechanism to modulate splicing from a variety of genetic contexts and demonstrate directly a novel therapeutic approach for SMA.


Subject(s)
Cyclic AMP Response Element-Binding Protein/genetics , Dependovirus/genetics , Genetic Vectors/genetics , Nerve Tissue Proteins/genetics , RNA-Binding Proteins/genetics , RNA/genetics , Blotting, Western , Cells, Cultured , Cyclic AMP Response Element-Binding Protein/metabolism , Exons/genetics , Fibroblasts/metabolism , Fibroblasts/pathology , HeLa Cells , Humans , Luciferases/genetics , Luciferases/metabolism , Microscopy, Fluorescence , Models, Genetic , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/pathology , Muscular Atrophy, Spinal/therapy , Nerve Tissue Proteins/metabolism , RNA/metabolism , RNA Precursors/genetics , RNA Precursors/metabolism , RNA Splicing/genetics , RNA-Binding Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , SMN Complex Proteins , Survival of Motor Neuron 1 Protein , Survival of Motor Neuron 2 Protein , Transfection/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...