Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
1.
Mol Ther Nucleic Acids ; 30: 17-27, 2022 Dec 13.
Article in English | MEDLINE | ID: mdl-36189424

ABSTRACT

Antisense RNA technology is a strategy for the treatment of Duchenne muscular dystrophy (DMD), a progressive and universally fatal X-linked neuromuscular disease caused by frameshift mutations in the gene encoding dystrophin. Phosphorodiamidate morpholino oligomers (PMOs) are an antisense RNA platform that is used clinically in patients with DMD to facilitate exon skipping and production of an internally truncated, yet functional, dystrophin protein. Peptide-conjugated PMOs (PPMOs) are a next-generation platform in which a cell-penetrating peptide is conjugated to the PMO backbone, with the goal of increasing cellular uptake. RC-1001 is a PPMO that contains a proprietary cell-penetrating peptide and targets the Dmd mutation in mdx mice. It was evaluated in mdx mice for exon 23 skipping, dystrophin production, and functional efficacy. Single-dose RC-1001 dose dependently increased exon skipping and dystrophin protein levels in striated muscle and is associated with improvements in muscle function. Dystrophin protein levels were durable for 60 days. Three doses, each given 1 month apart, increased exon skipping to 99% in quadriceps and 43% in heart, with dystrophin protein levels at 39% and 9% of wild type, respectively. These findings support clinical development of PPMO therapies for the treatment of DMD.

2.
Hum Mol Genet ; 23(23): 6318-31, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25055867

ABSTRACT

Reduced expression of SMN protein causes spinal muscular atrophy (SMA), a neurodegenerative disorder leading to motor neuron dysfunction and loss. However, the molecular mechanisms by which SMN regulates neuronal dysfunction are not fully understood. Here, we report that reduced SMN protein level alters miRNA expression and distribution in neurons. In particular, miR-183 levels are increased in neurites of SMN-deficient neurons. We demonstrate that miR-183 regulates translation of mTor via direct binding to its 3' UTR. Interestingly, local axonal translation of mTor is reduced in SMN-deficient neurons, and this can be recovered by miR-183 inhibition. Finally, inhibition of miR-183 expression in the spinal cord of an SMA mouse model prolongs survival and improves motor function of Smn-mutant mice. Together, these observations suggest that axonal miRNAs and the mTOR pathway are previously unidentified molecular mechanisms contributing to SMA pathology.


Subject(s)
Axons/metabolism , MicroRNAs/metabolism , Protein Biosynthesis , Survival of Motor Neuron 1 Protein/metabolism , TOR Serine-Threonine Kinases/biosynthesis , 3' Untranslated Regions , Animals , MicroRNAs/genetics , Muscular Atrophy, Spinal/metabolism , Muscular Atrophy, Spinal/pathology , Neurons/metabolism , Primary Cell Culture , RNA, Messenger/metabolism , Rats, Sprague-Dawley , Survival of Motor Neuron 1 Protein/genetics , TOR Serine-Threonine Kinases/genetics
3.
Mol Ther ; 22(8): 1450-1459, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24814151

ABSTRACT

Spinal muscular atrophy is a progressive motor neuron disease caused by a deficiency of survival motor neuron. In this study, we evaluated the efficacy of intravenous administration of a recombinant adeno-associated virus (AAV1) vector encoding human insulin-like growth factor-1 (IGF-1) in a severe mouse model of spinal muscular atrophy. Measurable quantities of human IGF-1 transcripts and protein were detected in the liver (up to 3 months postinjection) and in the serum indicating that IGF-1 was secreted from the liver into systemic circulation. Spinal muscular atrophy mice administered AAV1-IGF-1 on postnatal day 1 exhibited a lower extent of motor neuron degeneration, cardiac and muscle atrophy as well as a greater extent of innervation at the neuromuscular junctions compared to untreated controls at day 8 posttreatment. Importantly, treatment with AAV1-IGF-1 prolonged the animals' lifespan, increased their body weights and improved their motor coordination. Quantitative polymerase chain reaction and western blot analyses showed that AAV1-mediated expression of IGF-1 led to an increase in survival motor neuron transcript and protein levels in the spinal cord, brain, muscles, and heart. These data indicate that systemically delivered AAV1-IGF-1 can correct several of the biochemical and behavioral deficits in spinal muscular atrophy mice through increasing tissue levels of survival motor neuron.


Subject(s)
Genetic Therapy/methods , Genetic Vectors/administration & dosage , Insulin-Like Growth Factor I/genetics , Muscular Atrophy, Spinal/physiopathology , Muscular Atrophy, Spinal/therapy , Animals , Dependovirus/genetics , Disease Models, Animal , Humans , Injections, Intravenous , Insulin-Like Growth Factor I/administration & dosage , Liver/metabolism , Mice , Muscular Atrophy, Spinal/blood , Muscular Atrophy, Spinal/genetics , Survival of Motor Neuron 1 Protein/genetics , Survival of Motor Neuron 1 Protein/metabolism , Treatment Outcome
4.
Hum Gene Ther ; 25(7): 619-30, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24617515

ABSTRACT

Spinal muscular atrophy (SMA) is a neuromuscular disease caused by mutations in survival motor neuron 1 (SMN1). Previously, we showed that central nervous system (CNS) delivery of an adeno-associated viral (AAV) vector encoding SMN1 produced significant improvements in survival in a mouse model of SMA. Here, we performed a dose-response study in SMA mice to determine the levels of SMN in the spinal cord necessary for efficacy, and measured the efficiency of motor neuron transduction in the spinal cord after intrathecal delivery in pigs and nonhuman primates (NHPs). CNS injections of 5e10, 1e10, and 1e9 genome copies (gc) of self-complementary AAV9 (scAAV9)-hSMN1 into SMA mice extended their survival from 17 to 153, 70, and 18 days, respectively. Spinal cords treated with 5e10, 1e10, and 1e9 gc showed that 70-170%, 30-100%, and 10-20% of wild-type levels of SMN were attained, respectively. Furthermore, detectable SMN expression in a minimum of 30% motor neurons correlated with efficacy. A comprehensive analysis showed that intrathecal delivery of 2.5e13 gc of scAAV9-GFP transduced 25-75% of the spinal cord motor neurons in NHPs. Thus, the extent of gene expression in motor neurons necessary to confer efficacy in SMA mice could be obtained in large-animal models, justifying the continual development of gene therapy for SMA.


Subject(s)
Dependovirus , Genetic Vectors/pharmacology , Injections, Spinal , Muscular Atrophy, Spinal/therapy , Protein Biosynthesis , Survival of Motor Neuron 1 Protein , Animals , Genetic Vectors/genetics , Genetic Vectors/metabolism , Mice , Mice, Knockout , Motor Neurons/metabolism , Motor Neurons/pathology , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/metabolism , Muscular Atrophy, Spinal/pathology , Spinal Cord/metabolism , Spinal Cord/pathology , Survival of Motor Neuron 1 Protein/biosynthesis , Survival of Motor Neuron 1 Protein/genetics , Swine
5.
Proc Natl Acad Sci U S A ; 110(9): 3537-42, 2013 Feb 26.
Article in English | MEDLINE | ID: mdl-23297226

ABSTRACT

Mutations of GBA1, the gene encoding glucocerebrosidase, represent a common genetic risk factor for developing the synucleinopathies Parkinson disease (PD) and dementia with Lewy bodies. PD patients with or without GBA1 mutations also exhibit lower enzymatic levels of glucocerebrosidase in the central nervous system (CNS), suggesting a possible link between the enzyme and the development of the disease. Previously, we have shown that early treatment with glucocerebrosidase can modulate α-synuclein aggregation in a presymptomatic mouse model of Gaucher-related synucleinopathy (Gba1(D409V/D409V)) and ameliorate the associated cognitive deficit. To probe this link further, we have now evaluated the efficacy of augmenting glucocerebrosidase activity in the CNS of symptomatic Gba1(D409V/D409V) mice and in a transgenic mouse model overexpressing A53T α-synuclein. Adeno-associated virus-mediated expression of glucocerebrosidase in the CNS of symptomatic Gba1(D409V/D409V) mice completely corrected the aberrant accumulation of the toxic lipid glucosylsphingosine and reduced the levels of ubiquitin, tau, and proteinase K-resistant α-synuclein aggregates. Importantly, hippocampal expression of glucocerebrosidase in Gba1(D409V/D409V) mice (starting at 4 or 12 mo of age) also reversed their cognitive impairment when examined using a novel object recognition test. Correspondingly, overexpression of glucocerebrosidase in the CNS of A53T α-synuclein mice reduced the levels of soluble α-synuclein, suggesting that increasing the glycosidase activity can modulate α-synuclein processing and may modulate the progression of α-synucleinopathies. Hence, increasing glucocerebrosidase activity in the CNS represents a potential therapeutic strategy for GBA1-related and non-GBA1-associated synucleinopathies, including PD.


Subject(s)
Brain/enzymology , Gaucher Disease/drug therapy , Gaucher Disease/enzymology , Glucosylceramidase/metabolism , Parkinsonian Disorders/drug therapy , Parkinsonian Disorders/enzymology , alpha-Synuclein/metabolism , Animals , Brain/pathology , Brain/physiopathology , Dependovirus/metabolism , Disease Models, Animal , Gaucher Disease/pathology , Gaucher Disease/physiopathology , Glucosylceramidase/administration & dosage , Glucosylceramidase/genetics , Glucosylceramidase/therapeutic use , Hippocampus/metabolism , Hippocampus/pathology , Hippocampus/physiopathology , Humans , Memory , Mice , Mice, Transgenic , Parkinsonian Disorders/physiopathology , Protein Structure, Quaternary , Psychosine/analogs & derivatives , Psychosine/metabolism , alpha-Synuclein/genetics , tau Proteins/chemistry , tau Proteins/metabolism
6.
Mol Ther ; 20(10): 1893-901, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22828503

ABSTRACT

Niemann-Pick disease Type A (NPA) is a neuronopathic lysosomal storage disease (LSD) caused by the loss of acid sphingomyelinase (ASM). The goals of the current study are to ascertain the levels of human ASM that are efficacious in ASM knockout (ASMKO) mice, and determine whether these levels can be attained in non-human primates (NHPs) using a multiple parenchymal injection strategy. Intracranial injections of different doses of AAV1-hASM in ASMKO mice demonstrated that only a small amount of enzyme (<0.5 mg hASM/g tissue) was sufficient to increase survival, and that increasing the amount of hASM did not enhance this survival benefit until a new threshold level of >10 mg hASM/g tissue was reached. In monkeys, injection of 12 tracts of AAV1-hASM resulted in efficacious levels of enzyme in broad regions of the brain that was aided, in part, by axonal transport of adeno-associated virus (AAV) and movement through the perivascular space. This study demonstrates that a combination cortical, subcortical, and cerebellar injection protocol could provide therapeutic levels of hASM to regions of the NHP brain that are highly affected in NPA patients. The information from this study might help design new AAV-mediated enzyme replacement protocols for NPA and other neuronopathic LSDs in future clinical trials.


Subject(s)
Genetic Therapy , Niemann-Pick Disease, Type A/therapy , Sphingomyelin Phosphodiesterase/deficiency , Animals , Brain/enzymology , Dependovirus/genetics , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Genetic Vectors/genetics , Injections , Macaca fascicularis , Male , Mice , Mice, Knockout , Niemann-Pick Disease, Type A/pathology , Primates/metabolism , Sphingomyelin Phosphodiesterase/genetics , Sphingomyelin Phosphodiesterase/metabolism
7.
Mol Ther ; 20(9): 1713-23, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22735381

ABSTRACT

Central nervous system (CNS)-directed gene therapy with recombinant adeno-associated virus (AAV) vectors has been used effectively to slow disease course in mouse models of several neurodegenerative diseases. However, these vectors were typically tested in mice without prior exposure to the virus, an immunological scenario unlikely to be duplicated in human patients. Here, we examined the impact of pre-existing immunity on AAV-mediated gene delivery to the CNS of normal and diseased mice. Antibody levels in brain tissue were determined to be 0.6% of the levels found in systemic circulation. As expected, transgene expression in brains of mice with relatively high serum antibody titers was reduced by 59-95%. However, transduction activity was unaffected in mice that harbored more clinically relevant antibody levels. Moreover, we also showed that markers of neuroinflammation (GFAP, Iba1, and CD3) and histopathology (hematoxylin and eosin (H&E)) were not enhanced in immune-primed mice (regardless of pre-existing antibody levels). Importantly, we also demonstrated in a mouse model of Niemann Pick Type A (NPA) disease that pre-existing immunity did not preclude either gene transfer to the CNS or alleviation of disease-associated neuropathology. These findings support the continued development of AAV-based therapies for the treatment of neurological disorders.


Subject(s)
Antibodies, Viral/immunology , Brain/immunology , Dependovirus/genetics , Genetic Therapy/methods , Niemann-Pick Disease, Type A/therapy , Adult , Animals , Antibodies, Viral/metabolism , Biomarkers/metabolism , Brain/metabolism , Dependovirus/immunology , Disease Models, Animal , Gene Transfer Techniques , Genetic Vectors , Humans , Immunization , Mice , Niemann-Pick Disease, Type A/genetics , Niemann-Pick Disease, Type A/immunology , Niemann-Pick Disease, Type A/metabolism , Transgenes
8.
Neurobiol Dis ; 45(1): 272-9, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21884794

ABSTRACT

The efficacy of administering a recombinant adeno-associated virus (AAV) vector encoding human IGF-1 (AAV2/1-hIGF-1) into the deep cerebellar nucleus (DCN) of a type III SMA mouse model was evaluated. High levels of IGF-1 transcripts and protein were detected in the spinal cord at 2 months post-injection demonstrating that axonal connections between the cerebellum and spinal cord were able to act as conduits for the viral vector and protein to the spinal cord. Mice treated with AAV2/1-hIGF-1 and analyzed 8 months later showed changes in endogenous Bax and Bcl-xl levels in spinal cord motor neurons that were consistent with IGF-1-mediated anti-apoptotic effects on motor neurons. However, although AAV2/1-hIGF-1 treatment reduced the extent of motor neuron cell death, the majority of rescued motor neurons were non-functional, as they lacked axons that innervated the muscles. Furthermore, treated SMA mice exhibited abnormal muscle fibers, aberrant neuromuscular junction structure, and impaired performance on motor function tests. These data indicate that although CNS-directed expression of IGF-1 could reduce motor neuron cell death, this did not translate to improvements in motor function in an adult mouse model of type III SMA.


Subject(s)
Cell Death/drug effects , Insulin-Like Growth Factor I/therapeutic use , Motor Activity/drug effects , Motor Neurons/drug effects , Muscular Atrophy, Spinal/therapy , Animals , Cell Death/physiology , Cerebellum/drug effects , Cerebellum/pathology , Cerebellum/physiopathology , Genetic Therapy , Genetic Vectors , Insulin-Like Growth Factor I/pharmacology , Mice , Motor Activity/physiology , Motor Neurons/pathology , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/pathology , Muscular Atrophy, Spinal/pathology , Muscular Atrophy, Spinal/physiopathology , Spinal Cord/drug effects , Spinal Cord/pathology , Spinal Cord/physiopathology
9.
Proc Natl Acad Sci U S A ; 108(29): 12101-6, 2011 Jul 19.
Article in English | MEDLINE | ID: mdl-21730160

ABSTRACT

Emerging genetic and clinical evidence suggests a link between Gaucher disease and the synucleinopathies Parkinson disease and dementia with Lewy bodies. Here, we provide evidence that a mouse model of Gaucher disease (Gba1(D409V/D409V)) exhibits characteristics of synucleinopathies, including progressive accumulation of proteinase K-resistant α-synuclein/ubiquitin aggregates in hippocampal neurons and a coincident memory deficit. Analysis of homozygous (Gba1(D409V/D409V)) and heterozygous (Gba1(D409V/+) and Gba1(+/-)) Gaucher mice indicated that these pathologies are a result of the combination of a loss of glucocerebrosidase activity and a toxic gain-of-function resulting from expression of the mutant enzyme. Importantly, adeno-associated virus-mediated expression of exogenous glucocerebrosidase injected into the hippocampus of Gba1(D409V/D409V) mice ameliorated both the histopathological and memory aberrations. The data support the contention that mutations in GBA1 can cause Parkinson disease-like α-synuclein pathology, and that rescuing brain glucocerebrosidase activity might represent a therapeutic strategy for GBA1-associated synucleinopathies.


Subject(s)
Gaucher Disease/pathology , Glucosylceramidase/metabolism , Hippocampus/enzymology , alpha-Synuclein/metabolism , Analysis of Variance , Animals , Blotting, Western , Dependovirus , Endopeptidase K/metabolism , Gaucher Disease/metabolism , Gene Transfer Techniques , Genetic Vectors/genetics , Glucosylceramidase/genetics , Hippocampus/cytology , Immunohistochemistry , Mice
10.
Sci Transl Med ; 3(72): 72ra18, 2011 Mar 02.
Article in English | MEDLINE | ID: mdl-21368223

ABSTRACT

Spinal muscular atrophy (SMA) is an autosomal recessive neuromuscular disorder caused by mutations in the SMN1 gene that result in a deficiency of SMN protein. One approach to treat SMA is to use antisense oligonucleotides (ASOs) to redirect the splicing of a paralogous gene, SMN2, to boost production of functional SMN. Injection of a 2'-O-2-methoxyethyl-modified ASO (ASO-10-27) into the cerebral lateral ventricles of mice with a severe form of SMA resulted in splice-mediated increases in SMN protein and in the number of motor neurons in the spinal cord, which led to improvements in muscle physiology, motor function and survival. Intrathecal infusion of ASO-10-27 into cynomolgus monkeys delivered putative therapeutic levels of the oligonucleotide to all regions of the spinal cord. These data demonstrate that central nervous system-directed ASO therapy is efficacious and that intrathecal infusion may represent a practical route for delivering this therapeutic in the clinic.


Subject(s)
Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/pathology , Muscular Atrophy, Spinal/therapy , Oligonucleotides, Antisense/therapeutic use , Spinal Cord/pathology , Animals , Disease Models, Animal , Drug Delivery Systems , Humans , Macaca fascicularis , Mice , Motor Neurons/physiology , Muscular Atrophy, Spinal/physiopathology , Neuromuscular Junction/ultrastructure , Oligonucleotides, Antisense/administration & dosage , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/pharmacokinetics , RNA Splicing , Spinal Cord/physiopathology , Survival of Motor Neuron 1 Protein/genetics , Survival of Motor Neuron 2 Protein/genetics
11.
Trends Mol Med ; 17(5): 259-65, 2011 May.
Article in English | MEDLINE | ID: mdl-21334976

ABSTRACT

Spinal muscular atrophy (SMA) is a neuromuscular disease caused by a deficiency of functional SMN protein because of mutations in SMN1. A decrease in SMN activity results in motor neuron cell loss in the spinal cord, leading to a weakness of the proximal muscles responsible for crawling, walking, head/neck control and swallowing as well as the involuntary muscles that control breathing and coughing. Thus, patients present with pulmonary manifestations, paralysis and a shortened lifespan. Gene therapy is emerging as a promising therapeutic strategy for SMA given that the molecular basis for this monogenic disorder is well established. Recent advances and findings from preclinical studies in animal models provide optimism that gene therapy might be an effective therapeutic strategy for treating SMA.


Subject(s)
Genetic Therapy , Muscular Atrophy, Spinal/therapy , Animals , Humans , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/metabolism , SMN Complex Proteins/metabolism , Spinal Cord/metabolism , Survival of Motor Neuron 1 Protein/genetics
12.
Genes Dev ; 24(15): 1634-44, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20624852

ABSTRACT

Increasing survival of motor neuron 2, centromeric (SMN2) exon 7 inclusion to express more full-length SMN protein in motor neurons is a promising approach to treat spinal muscular atrophy (SMA), a genetic neurodegenerative disease. Previously, we identified a potent 2'-O-(2-methoxyethyl) (MOE) phosphorothioate-modified antisense oligonucleotide (ASO) that blocks an SMN2 intronic splicing silencer element and efficiently promotes exon 7 inclusion in transgenic mouse peripheral tissues after systemic administration. Here we address its efficacy in the spinal cord--a prerequisite for disease treatment--and its ability to rescue a mild SMA mouse model that develops tail and ear necrosis, resembling the distal tissue necrosis reported in some SMA infants. Using a micro-osmotic pump, we directly infused the ASO into a lateral cerebral ventricle in adult mice expressing a human SMN2 transgene; the ASO gave a robust and long-lasting increase in SMN2 exon 7 inclusion measured at both the mRNA and protein levels in spinal cord motor neurons. A single embryonic or neonatal intracerebroventricular ASO injection strikingly rescued the tail and ear necrosis in SMA mice. We conclude that this MOE ASO is a promising drug candidate for SMA therapy, and, more generally, that ASOs can be used to efficiently redirect alternative splicing of target genes in the CNS.


Subject(s)
Alternative Splicing , Motor Neurons/drug effects , Muscular Atrophy, Spinal , Necrosis/physiopathology , Oligonucleotides, Antisense/pharmacology , Survival of Motor Neuron 2 Protein/genetics , Survival of Motor Neuron 2 Protein/metabolism , Animals , Animals, Newborn , Disease Models, Animal , Embryo, Mammalian , Female , Gene Expression Regulation/drug effects , Male , Mice , Muscular Atrophy, Spinal/physiopathology , Muscular Atrophy, Spinal/therapy , Necrosis/drug therapy , Oligonucleotides, Antisense/administration & dosage , Oligonucleotides, Antisense/pharmacokinetics , Spinal Cord/drug effects , Spinal Cord/metabolism , Transgenes/genetics
13.
J Clin Invest ; 120(4): 1253-64, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20234094

ABSTRACT

Spinal muscular atrophy (SMA) is a neuromuscular disease caused by a deficiency of survival motor neuron (SMN) due to mutations in the SMN1 gene. In this study, an adeno-associated virus (AAV) vector expressing human SMN (AAV8-hSMN) was injected at birth into the CNS of mice modeling SMA. Western blot analysis showed that these injections resulted in widespread expression of SMN throughout the spinal cord, and this translated into robust improvement in skeletal muscle physiology, including increased myofiber size and improved neuromuscular junction architecture. Treated mice also displayed substantial improvements on behavioral tests of muscle strength, coordination, and locomotion, indicating that the neuromuscular junction was functional. Treatment with AAV8-hSMN increased the median life span of mice with SMA-like disease to 50 days compared with 15 days for untreated controls. Moreover, injecting mice with SMA-like disease with a human SMN-expressing self-complementary AAV vector - a vector that leads to earlier onset of gene expression compared with standard AAV vectors - led to improved efficacy of gene therapy, including a substantial extension in median survival to 157 days. These data indicate that CNS-directed, AAV-mediated SMN augmentation is highly efficacious in addressing both neuronal and muscular pathologies in a severe mouse model of SMA.


Subject(s)
Genetic Therapy , Motor Neurons/physiology , Muscular Atrophy, Spinal/therapy , Survival of Motor Neuron 1 Protein/genetics , Animals , Disease Models, Animal , Humans , Mice , Muscle Strength , Muscle, Skeletal/pathology , Muscular Atrophy, Spinal/mortality , Muscular Atrophy, Spinal/physiopathology , Neurites/metabolism , Neuromuscular Junction/pathology
14.
Exp Neurol ; 215(2): 349-57, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19059399

ABSTRACT

Niemann-Pick A (NPA) disease is a lysosomal storage disorder (LSD) caused by a deficiency in acid sphingomyelinase (ASM) activity. Previously, we showed that the storage pathology in the ASM knockout (ASMKO) mouse brain could be corrected by intracerebral injections of cell, gene and protein based therapies. However, except for instances where distal areas were targeted with viral vectors, correction of lysosomal storage pathology was typically limited to a region within a few millimeters from the injection site. As NPA is a global neurometabolic disease, the development of delivery strategies that maximize the distribution of the enzyme throughout the CNS is likely necessary to arrest or delay progression of the disease. To address this challenge, we evaluated the effectiveness of intracerebroventricular (ICV) delivery of recombinant human ASM into ASMKO mice. Our findings showed that ICV delivery of the enzyme led to widespread distribution of the hydrolase throughout the CNS. Moreover, a significant reduction in lysosomal accumulation of sphingomyelin was observed throughout the brain and also within the spinal cord and viscera. Importantly, we demonstrated that repeated ICV infusions of ASM were effective at improving the disease phenotype in the ASMKO mouse as indicated by a partial alleviation of the motor abnormalities. These findings support the continued exploration of ICV delivery of recombinant lysosomal enzymes as a therapeutic modality for LSDs such as NPA that manifests substrate accumulation within the CNS.


Subject(s)
Niemann-Pick Disease, Type A/drug therapy , Sphingomyelin Phosphodiesterase/administration & dosage , Animals , Brain/metabolism , Cholesterol/metabolism , Disease Models, Animal , Humans , Injections, Intraventricular/methods , Lysosomes/drug effects , Lysosomes/metabolism , Mice , Mice, Knockout , Niemann-Pick Disease, Type A/genetics , Niemann-Pick Disease, Type A/pathology , Sphingomyelin Phosphodiesterase/deficiency , Sphingomyelin Phosphodiesterase/metabolism , Sphingomyelins/metabolism , Time Factors
15.
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
16.
J Neuropathol Exp Neurol ; 67(8): 803-18, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18648322

ABSTRACT

Pompe disease (glycogen storage disease II) is caused by mutations in the acid alpha-glucosidase gene. The most common form is rapidly progressive with glycogen storage, particularly in muscle, which leads to profound weakness, cardiac failure, and death by the age of 2 years. Although usually considered a muscle disease, glycogen storage also occurs in the CNS. We evaluated the progression of neuropathologic and behavioral abnormalities in a Pompe disease mouse model (6neo/6neo) that displays many features of the human disease. Homozygous mutant mice store excess glycogen within large neurons of hindbrain, spinal cord, and sensory ganglia by the age of 1 month; accumulations then spread progressively within many CNS cell types. "Silver degeneration" and Fluoro-Jade C stains revealed severe degeneration in axon terminals of primary sensory neurons at 3 to 9 months. These abnormalities were accompanied by progressive behavioral impairment on rotorod, wire hanging, and foot fault tests. The extensive neuropathologic alterations in this model suggest that therapy of skeletal and cardiac muscle disorders by systemic enzyme replacement therapy may not be sufficient to reverse functional deficits due to CNS glycogen storage, particularly early-onset, rapidly progressive disease. A better understanding of the basis for clinical manifestations is needed to correlate CNS pathology with Pompe disease manifestations.


Subject(s)
Behavior, Animal/physiology , Central Nervous System/pathology , Disease Models, Animal , Glycogen Storage Disease Type II/pathology , Glycogen Storage Disease Type II/physiopathology , Phenotype , Age Factors , Animals , Central Nervous System/ultrastructure , Disease Progression , Glial Fibrillary Acidic Protein/metabolism , Glycogen/metabolism , Glycogen Storage Disease Type II/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Transmission/methods , Motor Activity/physiology , Muscle Strength/physiology , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Psychomotor Performance/physiology , Reaction Time/physiology , alpha-Glucosidases/deficiency
17.
Mol Ther ; 16(6): 1056-64, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18388910

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease of the motor system. Recent work in rodent models of ALS has shown that insulin-like growth factor-1 (IGF-1) slows disease progression when delivered at disease onset. However, IGF-1's mechanism of action along the neuromuscular axis remains unclear. In this study, symptomatic ALS mice received IGF-1 through stereotaxic injection of an IGF-1-expressing viral vector to the deep cerebellar nuclei (DCN), a region of the cerebellum with extensive brain stem and spinal cord connections. We found that delivery of IGF-1 to the central nervous system (CNS) reduced ALS neuropathology, improved muscle strength, and significantly extended life span in ALS mice. To explore the mechanism of action of IGF-1, we used a newly developed in vitro model of ALS. We demonstrate that IGF-1 is potently neuroprotective and attenuates glial cell-mediated release of tumor necrosis factor-alpha (TNF-alpha) and nitric oxide (NO). Our results show that delivering IGF-1 to the CNS is sufficient to delay disease progression in a mouse model of familial ALS and demonstrate for the first time that IGF-1 attenuates the pathological activity of non-neuronal cells that contribute to disease progression. Our findings highlight an innovative approach for delivering IGF-1 to the CNS.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/therapy , Central Nervous System/cytology , Dependovirus/genetics , Genetic Therapy/methods , Insulin-Like Growth Factor I/genetics , Neuroglia/cytology , Neuroglia/metabolism , Animals , Cell Survival , Central Nervous System/metabolism , Cerebellum/metabolism , Female , Insulin-Like Growth Factor I/metabolism , Male , Mice , Neurodegenerative Diseases/metabolism , Tumor Necrosis Factor-alpha/metabolism
18.
Mol Ther ; 16(4): 649-56, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18362923

ABSTRACT

Late infantile neuronal ceroid lipofuscinosis (LINCL) is an autosomal recessive neurodegenerative disease caused by mutations in CLN2, which encodes the lysosomal protease tripeptidyl peptidase 1 (TPP1). LINCL is characterized clinically by progressive motor and cognitive decline, and premature death. Enzyme-replacement therapy (ERT) is currently available for lysosomal storage diseases affecting peripheral tissues, but has not been used in patients with central nervous system (CNS) involvement. Enzyme delivery through the cerebrospinal fluid is a potential alternative route to the CNS, but has not been studied for LINCL. In this study, we identified relevant neuropathological and behavioral hallmarks of disease in a mouse model of LINCL and correlated those findings with tissues from LINCL patients. Subsequently, we tested if intraventricular delivery of TPP1 to the LINCL mouse was efficacious. We found that infusion of recombinant human TPP1 through an intraventricular cannula led to enzyme distribution in several regions of the brain of treated mice. In vitro activity assays confirm increased TPP1 activity throughout the rostral-caudal extent of the brain. Importantly, treated mice showed attenuated neuropathology, and decreased resting tremor relative to vehicle-treated mice. This data demonstrates that intraventricular enzyme delivery to the CNS is feasible and may be of therapeutic value.


Subject(s)
Endopeptidases/therapeutic use , Neuronal Ceroid-Lipofuscinoses/therapy , Adult , Aminopeptidases , Animals , Astrocytes/metabolism , Brain/enzymology , Brain/pathology , Cerebral Ventricles , Child , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases , Disease Models, Animal , Endopeptidases/genetics , Endopeptidases/metabolism , Glial Fibrillary Acidic Protein/metabolism , Humans , Infant , Mice , Mice, Knockout , Neuronal Ceroid-Lipofuscinoses/pathology , Neuronal Ceroid-Lipofuscinoses/physiopathology , Neurons/metabolism , Phenotype , Purkinje Cells/metabolism , Purkinje Cells/pathology , Recombinant Proteins/therapeutic use , Serine Proteases , Tripeptidyl-Peptidase 1
19.
Exp Neurol ; 207(2): 258-66, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17686472

ABSTRACT

Niemann-Pick A disease (NPD-A) is caused by a deficiency of acid sphingomyelinase (ASM) leading to the intracellular accumulation of sphingomyelin and cholesterol in lysosomes. We evaluated the effects of direct intraparenchymal brain injections of purified recombinant human ASM (hASM) at correcting the storage pathology in a mouse model of NPD-A (ASMKO). Different doses (0.1 ng to 10 mug of hASM) were injected into the right hemisphere of the hippocampus and thalamus of 12- to 14-week-old ASMKO mice. Immunohistochemical analysis after 1 week indicated that animals treated with greater than 1 mug hASM/site showed detectable levels of enzyme around the injected regions. However, localized clearance of sphingomyelin and cholesterol storage were observed in animals administered lower doses of enzyme, starting at 100 ng hASM/site. Areas of correction were also noted at distal sites such as in the contralateral hemispheres. Indications of storage re-accumulation were seen after 2 weeks post-injection. Injections of hASM did not cause any significant cell infiltration, astrogliosis, or microglial activation. These results indicate that intraparenchymal injection of hASM is associated with minimal toxicity and can lead to regional reductions in storage pathology in the ASMKO mouse.


Subject(s)
Lysosomes/metabolism , Niemann-Pick Disease, Type A/drug therapy , Niemann-Pick Disease, Type A/pathology , Sphingomyelin Phosphodiesterase/therapeutic use , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Administration Routes , Filipin/metabolism , Mice , Mice, Knockout , Niemann-Pick Disease, Type A/genetics , Sphingomyelin Phosphodiesterase/deficiency , Time Factors , Toxins, Biological/metabolism
20.
Mol Ther ; 15(10): 1782-8, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17637720

ABSTRACT

Classical late infantile neuronal ceroid lipofuscinosis (cLINCL) is a monogenic disorder caused by the loss of tripeptidyl peptidase 1 (TPP1) activity as a result of mutations in CLN2. Absence of TPP1 results in lysosomal storage with an accompanying axonal degeneration throughout the central nervous system (CNS), which leads to progressive neurodegeneration and early death. In this study, we compared the efficacies of pre- and post-symptomatic injections of recombinant adeno-associated virus (AAV) for treating the cellular and functional abnormalities of CLN2 mutant mice. Intracranial injection of AAV1-hCLN2 resulted in widespread human TPP1 (hTPP1) activity in the brain that was 10-100-fold above wild-type levels. Injections before disease onset prevented storage and spared neurons from axonal degeneration, reflected by the preservation of motor function. Furthermore, the majority of CLN2 mutant mice treated pre-symptomatically lived for at least 330 days, compared with a median survival of 151 days in untreated CLN2 mutant controls. In contrast, although injection after disease onset ameliorated lysosomal storage, there was evidence of axonal degeneration, motor function showed limited recovery, and the animals had a median lifespan of 216 days. These data illustrate the importance of early intervention for enhanced therapeutic benefit, which may provide guidance in designing novel treatment strategies for cLINCL patients.


Subject(s)
Disease Models, Animal , Neuronal Ceroid-Lipofuscinoses/therapy , Aminopeptidases , Animals , Brain/pathology , Dependovirus/genetics , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases , Endopeptidases/genetics , Genetic Therapy , Genetic Vectors , Mice , Mice, Mutant Strains , Motor Activity , Neuronal Ceroid-Lipofuscinoses/physiopathology , Serine Proteases , Survival Analysis , Tripeptidyl-Peptidase 1
SELECTION OF CITATIONS
SEARCH DETAIL
...