Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
2.
N Engl J Med ; 377(18): 1713-1722, 2017 11 02.
Article in English | MEDLINE | ID: mdl-29091557

ABSTRACT

BACKGROUND: Spinal muscular atrophy type 1 (SMA1) is a progressive, monogenic motor neuron disease with an onset during infancy that results in failure to achieve motor milestones and in death or the need for mechanical ventilation by 2 years of age. We studied functional replacement of the mutated gene encoding survival motor neuron 1 (SMN1) in this disease. METHODS: Fifteen patients with SMA1 received a single dose of intravenous adeno-associated virus serotype 9 carrying SMN complementary DNA encoding the missing SMN protein. Three of the patients received a low dose (6.7×1013 vg per kilogram of body weight), and 12 received a high dose (2.0×1014 vg per kilogram). The primary outcome was safety. The secondary outcome was the time until death or the need for permanent ventilatory assistance. In exploratory analyses, we compared scores on the CHOP INTEND (Children's Hospital of Philadelphia Infant Test of Neuromuscular Disorders) scale of motor function (ranging from 0 to 64, with higher scores indicating better function) in the two cohorts and motor milestones in the high-dose cohort with scores in studies of the natural history of the disease (historical cohorts). RESULTS: As of the data cutoff on August 7, 2017, all 15 patients were alive and event-free at 20 months of age, as compared with a rate of survival of 8% in a historical cohort. In the high-dose cohort, a rapid increase from baseline in the score on the CHOP INTEND scale followed gene delivery, with an increase of 9.8 points at 1 month and 15.4 points at 3 months, as compared with a decline in this score in a historical cohort. Of the 12 patients who had received the high dose, 11 sat unassisted, 9 rolled over, 11 fed orally and could speak, and 2 walked independently. Elevated serum aminotransferase levels occurred in 4 patients and were attenuated by prednisolone. CONCLUSIONS: In patients with SMA1, a single intravenous infusion of adeno-associated viral vector containing DNA coding for SMN resulted in longer survival, superior achievement of motor milestones, and better motor function than in historical cohorts. Further studies are necessary to confirm the safety and efficacy of this gene therapy. (Funded by AveXis and others; ClinicalTrials.gov number, NCT02122952 .).


Subject(s)
Genetic Therapy , Spinal Muscular Atrophies of Childhood/therapy , Survival of Motor Neuron 1 Protein/genetics , Cohort Studies , Dependovirus , Disease-Free Survival , Female , Genetic Therapy/adverse effects , Genetic Vectors , Historically Controlled Study , Humans , Infant , Infant, Newborn , Infusions, Intravenous , Liver Diseases/etiology , Male , Motor Skills , Nutritional Support , Respiration, Artificial , Spinal Muscular Atrophies of Childhood/genetics , Spinal Muscular Atrophies of Childhood/physiopathology
3.
Ann Clin Transl Neurol ; 4(2): 76-86, 2017 02.
Article in English | MEDLINE | ID: mdl-28168207

ABSTRACT

OBJECTIVE: Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease in which glia are central mediators of motor neuron (MN) death. Since multiple cell types are involved in disease pathogenesis, the objective of this study was to determine the benefit of co-targeting independent pathogenic mechanisms in a familial ALS mouse model. METHODS: Recently, our laboratory identified that ALS microglia induce MN death in an NF-κB-dependent mechanism. We also demonstrated that a single, post-natal, intravenous injection of adeno-associated viral vector serotype 9 encoding a shRNA against mutant SOD1 is able to traverse the blood-brain barrier of ALS mice and reduce SOD1-expression in astrocytes and MNs. Reducing mutant SOD1 in MNs and astrocytes led to a robust increase in survival. To evaluate the benefit of co-targeting multiple cell types in ALS, we combined microglial NF-κB suppression with SOD1 reduction in astrocytes and MNs. RESULTS: Targeting both astrocytes and microglia resulted in an additive increase in survival and motor function by delaying both onset and progression. Strikingly, targeting all three cell types (astrocytes, motor neurons [MNs], and microglia) resulted in an additive increase in lifespan and motor function, with maximum survival reaching 204 days, 67 days longer than the mean survival of untreated control animals. INTERPRETATION: Our data suggest that a combinatorial approach co-targeting different pathogenic mechanisms in independent cell types is a beneficial therapeutic strategy for ALS.

4.
Proc Natl Acad Sci U S A ; 113(42): E6496-E6505, 2016 10 18.
Article in English | MEDLINE | ID: mdl-27688759

ABSTRACT

Oligodendrocytes have recently been implicated in the pathophysiology of amyotrophic lateral sclerosis (ALS). Here we show that, in vitro, mutant superoxide dismutase 1 (SOD1) mouse oligodendrocytes induce WT motor neuron (MN) hyperexcitability and death. Moreover, we efficiently derived human oligodendrocytes from a large number of controls and patients with sporadic and familial ALS, using two different reprogramming methods. All ALS oligodendrocyte lines induced MN death through conditioned medium (CM) and in coculture. CM-mediated MN death was associated with decreased lactate production and release, whereas toxicity in coculture was lactate-independent, demonstrating that MN survival is mediated not only by soluble factors. Remarkably, human SOD1 shRNA treatment resulted in MN rescue in both mouse and human cultures when knockdown was achieved in progenitor cells, whereas it was ineffective in differentiated oligodendrocytes. In fact, early SOD1 knockdown rescued lactate impairment and cell toxicity in all lines tested, with the exclusion of samples carrying chromosome 9 ORF 72 (C9orf72) repeat expansions. These did not respond to SOD1 knockdown nor did they show lactate release impairment. Our data indicate that SOD1 is directly or indirectly involved in ALS oligodendrocyte pathology and suggest that in this cell type, some damage might be irreversible. In addition, we demonstrate that patients with C9ORF72 represent an independent patient group that might not respond to the same treatment.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Motor Neurons/metabolism , Oligodendroglia/metabolism , Superoxide Dismutase-1/genetics , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Animals , Apoptosis , Biomarkers , C9orf72 Protein/genetics , C9orf72 Protein/metabolism , Cell Communication , Cell Death , Cell Differentiation , Cell Survival , Disease Models, Animal , Gene Expression Profiling , Humans , Lactic Acid/metabolism , Mice , Mice, Transgenic , Mutation , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Oligodendroglia/cytology , Superoxide Dismutase-1/metabolism
5.
Thyroid ; 26(9): 1311-9, 2016 09.
Article in English | MEDLINE | ID: mdl-27432638

ABSTRACT

BACKGROUND: MCT8 gene mutations produce thyroid hormone (TH) deficiency in the brain, causing severe neuropsychomotor abnormalities not correctable by treatment with TH. This proof-of-concept study examined whether transfer of human MCT8 (hMCT8) cDNA using adeno-associated virus 9 (AAV9) could correct the brain defects of Mct8 knockout mice (Mct8KO). METHODS: AAV9 vectors delivering long and/or short hMCT8 protein isoforms or an empty vector were injected intravenously (IV) and/or intracerebroventricularly (ICV) into postnatal day 1 Mct8KO and wild type (Wt) mice. Triiodothyronine (T3) was given daily for four days before postnatal day 28, at which time brains were collected after perfusion to assess increase in T3 content and effect on the T3-responsive transcription factor, Hairless. RESULTS: Increased pup mortality was observed after IV injection of the AAV9-long hMCT8 isoform, but not after injection of AAV9-short hMCT8 isoform. Compared to IV, ICV delivery produced more hMCT8 mRNA and protein relative to the viral dose, which was present in various brain regions and localized to the cell membranes. Despite production of abundant hMCT8 mRNA and protein with ICV delivery, only IV delivered AAV9-hMCT8 targeted the choroid plexus and significantly increased brain T3 content and expression of Hairless. CONCLUSIONS: These results indicate that MCT8 delivery to brain barriers by IV but not ICV injection is crucial for its proper function. MCT8 has no constitutive activity but acts through an increase in T3 entering the brain tissue. Increasing MCT8 expression in brain cell membranes, including neurons, is insufficient to produce an effect without an increase in brain T3 content. The correct hMCT8 isoform along with an optimized delivery method are critical for an effective gene therapy to provide functional MCT8 in the brain of patients with MCT8 mutations.


Subject(s)
Brain/metabolism , Membrane Transport Proteins/genetics , Thyroid Hormones/metabolism , Animals , Brain/drug effects , Dependovirus , Genetic Therapy , Membrane Transport Proteins/metabolism , Mice , Mice, Knockout , Monocarboxylic Acid Transporters , Symporters , Thyroid Hormones/blood , Triiodothyronine/pharmacology
6.
Nat Med ; 22(4): 397-403, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26928464

ABSTRACT

Astrocytes isolated from individuals with amyotrophic lateral sclerosis (ALS) are toxic to motor neurons (MNs) and play a non-cell autonomous role in disease pathogenesis. The mechanisms underlying the susceptibility of MNs to cell death remain unclear. Here we report that astrocytes derived from either mice bearing mutations in genes associated with ALS or human subjects with ALS reduce the expression of major histocompatibility complex class I (MHCI) molecules on MNs; reduced MHCI expression makes these MNs susceptible to astrocyte-induced cell death. Increasing MHCI expression on MNs increases survival and motor performance in a mouse model of ALS and protects MNs against astrocyte toxicity. Overexpression of a single MHCI molecule, HLA-F, protects human MNs from ALS astrocyte-mediated toxicity, whereas knockdown of its receptor, the killer cell immunoglobulin-like receptor KIR3DL2, on human astrocytes results in enhanced MN death. Thus, our data indicate that, in ALS, loss of MHCI expression on MNs renders them more vulnerable to astrocyte-mediated toxicity.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Histocompatibility Antigens Class I/biosynthesis , Motor Neurons/pathology , Receptors, KIR3DL2/genetics , Aged , Aged, 80 and over , Amyotrophic Lateral Sclerosis/pathology , Animals , Astrocytes/metabolism , Astrocytes/pathology , Cadaver , Cell Death/genetics , Disease Models, Animal , Female , Gene Expression Regulation , Histocompatibility Antigens Class I/genetics , Humans , Male , Mice , Mice, Transgenic , Middle Aged , Mutation , Superoxide Dismutase/genetics
7.
Mol Ther ; 23(1): 192-201, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25322757

ABSTRACT

Becker muscular dystrophy (BMD) is a variant of dystrophin deficiency resulting from DMD gene mutations. Phenotype is variable with loss of ambulation in late teenage or late mid-life years. There is currently no treatment for this condition. In this BMD proof-of-principle clinical trial, a potent myostatin antagonist, follistatin (FS), was used to inhibit the myostatin pathway. Extensive preclinical studies, using adeno-associated virus (AAV) to deliver follistatin, demonstrated an increase in strength. For this trial, we used the alternatively spliced FS344 to avoid potential binding to off target sites. AAV1.CMV.FS344 was delivered to six BMD patients by direct bilateral intramuscular quadriceps injections. Cohort 1 included three subjects receiving 3 × 10(11) vg/kg/leg. The distance walked on the 6MWT was the primary outcome measure. Patients 01 and 02 improved 58 meters (m) and 125 m, respectively. Patient 03 showed no change. In Cohort 2, Patients 05 and 06 received 6 × 10(11) vg/kg/leg with improved 6MWT by 108 m and 29 m, whereas, Patient 04 showed no improvement. No adverse effects were encountered. Histological changes corroborated benefit showing reduced endomysial fibrosis, reduced central nucleation, more normal fiber size distribution with muscle hypertrophy, especially at high dose. The results are encouraging for treatment of dystrophin-deficient muscle diseases.


Subject(s)
Dystrophin/deficiency , Follistatin-Related Proteins/genetics , Genetic Therapy/methods , Muscular Dystrophy, Duchenne/therapy , Myostatin/genetics , Adult , Dependovirus/genetics , Dystrophin/genetics , Follistatin-Related Proteins/metabolism , Gene Expression , Genetic Vectors , Humans , Injections, Intramuscular , Male , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/pathology , Myostatin/antagonists & inhibitors , Myostatin/metabolism
8.
Mol Ther ; 23(3): 477-87, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25358252

ABSTRACT

Spinal muscular atrophy (SMA) is the most frequent lethal genetic neurodegenerative disorder in infants. The disease is caused by low abundance of the survival of motor neuron (SMN) protein leading to motor neuron degeneration and progressive paralysis. We previously demonstrated that a single intravenous injection (IV) of self-complementary adeno-associated virus-9 carrying the human SMN cDNA (scAAV9-SMN) resulted in widespread transgene expression in spinal cord motor neurons in SMA mice as well as nonhuman primates and complete rescue of the disease phenotype in mice. Here, we evaluated the dosing and efficacy of scAAV9-SMN delivered directly to the cerebral spinal fluid (CSF) via single injection. We found widespread transgene expression throughout the spinal cord in mice and nonhuman primates when using a 10 times lower dose compared to the IV application. Interestingly, in nonhuman primates, lower doses than in mice can be used for similar motor neuron targeting efficiency. Moreover, the transduction efficacy is further improved when subjects are kept in the Trendelenburg position to facilitate spreading of the vector. We present a detailed analysis of transduction levels throughout the brain, brainstem, and spinal cord of nonhuman primates, providing new guidance for translation toward therapy for a wide range of neurodegenerative disorders.


Subject(s)
Dependovirus/genetics , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Muscular Atrophy, Spinal/therapy , Spinal Cord/metabolism , Survival of Motor Neuron 1 Protein/genetics , Animals , Animals, Newborn , Brain Stem/metabolism , Cerebral Cortex/metabolism , DNA, Complementary/administration & dosage , DNA, Complementary/genetics , DNA, Complementary/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Gene Expression , Genetic Vectors/pharmacokinetics , Injections, Epidural , Macaca fascicularis , Mice , Mice, Knockout , Motor Neurons/metabolism , Motor Neurons/pathology , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/metabolism , Muscular Atrophy, Spinal/pathology , Spinal Cord/pathology , Survival of Motor Neuron 1 Protein/metabolism , Transduction, Genetic , Transgenes
9.
Neuron ; 81(5): 1009-1023, 2014 Mar 05.
Article in English | MEDLINE | ID: mdl-24607225

ABSTRACT

Neuroinflammation is one of the most striking hallmarks of amyotrophic lateral sclerosis (ALS). Nuclear factor-kappa B (NF-κB), a master regulator of inflammation, is upregulated in spinal cords of ALS patients and SOD1-G93A mice. In this study, we show that selective NF-κB inhibition in ALS astrocytes is not sufficient to rescue motor neuron (MN) death. However, the localization of NF-κB activity and subsequent deletion of NF-κB signaling in microglia rescued MNs from microglial-mediated death in vitro and extended survival in ALS mice by impairing proinflammatory microglial activation. Conversely, constitutive activation of NF-κB selectively in wild-type microglia induced gliosis and MN death in vitro and in vivo. Taken together, these data provide a mechanism by which microglia induce MN death in ALS and suggest a novel therapeutic target that can be modulated to slow the progression of ALS and possibly other neurodegenerative diseases by which microglial activation plays a role.


Subject(s)
Amyotrophic Lateral Sclerosis/pathology , Cell Death/physiology , Microglia/cytology , Motor Neurons/cytology , NF-kappa B/metabolism , Age Factors , Amyotrophic Lateral Sclerosis/metabolism , Animals , Animals, Newborn , Astrocytes/cytology , Astrocytes/metabolism , Cell Communication/physiology , Coculture Techniques , Disease Models, Animal , Female , Mice , Mice, Inbred Strains , Mice, Transgenic , Microglia/metabolism , Motor Neurons/metabolism , NF-kappa B/antagonists & inhibitors , Primary Cell Culture , Signal Transduction/physiology , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Superoxide Dismutase-1
10.
Mol Ther ; 21(12): 2148-59, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24008656

ABSTRACT

Mutations in superoxide dismutase 1 (SOD1) are linked to familial amyotrophic lateral sclerosis (ALS) resulting in progressive motor neuron death through one or more acquired toxicities. Involvement of wild-type SOD1 has been linked to sporadic ALS, as misfolded SOD1 has been reported in affected tissues of sporadic patients and toxicity of astrocytes derived from sporadic ALS patients to motor neurons has been reported to be reduced by lowering the synthesis of SOD1. We now report slowed disease onset and progression in two mouse models following therapeutic delivery using a single peripheral injection of an adeno-associated virus serotype 9 (AAV9) encoding an shRNA to reduce the synthesis of ALS-causing human SOD1 mutants. Delivery to young mice that develop aggressive, fatal paralysis extended survival by delaying both disease onset and slowing progression. In a later-onset model, AAV9 delivery after onset markedly slowed disease progression and significantly extended survival. Moreover, AAV9 delivered intrathecally to nonhuman primates is demonstrated to yield robust SOD1 suppression in motor neurons and glia throughout the spinal cord and therefore, setting the stage for AAV9-mediated therapy in human clinical trials.


Subject(s)
Amyotrophic Lateral Sclerosis/pathology , Amyotrophic Lateral Sclerosis/therapy , Dependovirus/genetics , Genetic Therapy , Motor Neurons/metabolism , Neuroglia/metabolism , RNA, Small Interfering/genetics , Superoxide Dismutase/genetics , Administration, Intravenous , Amyotrophic Lateral Sclerosis/genetics , Animals , COS Cells , Chlorocebus aethiops , Disease Models, Animal , Disease Progression , Female , Genetic Vectors , HEK293 Cells , Humans , Injections, Spinal , Macaca fascicularis , Mice , Motor Neurons/pathology , Neuroglia/pathology , Superoxide Dismutase/metabolism , Superoxide Dismutase-1
11.
Aging Cell ; 11(3): 542-52, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22404871

ABSTRACT

Accumulating evidence suggests that adult hippocampal neurogenesis relies on the controlled and continued proliferation of neural progenitor cells (NPCs). With age, neurogenesis decreases through mechanisms that remain unclear but are believed to involve changes in the NPC microenvironment. Here, we provide evidence that NPC proliferation in the adult brain is in part regulated by astrocytes via Wnt signaling and that this cellular cross-talk is modified in the aging brain, leading to decreased proliferation of NPCs. Furthermore, we show that astrocytes regulate the NPC cell cycle by acting on the expression levels of survivin, a known mitotic regulator. Among cell cycle genes found down-regulated in aged NPCs, survivin was the only one that restored NPC proliferation in the aged brain. Our results provide a mechanism for the gradual loss of neurogenesis in the brain associated with aging and suggest that targeted modulation of survivin expression directly or through Wnt signaling could be used to stimulate adult neurogenesis.


Subject(s)
Hippocampus/cytology , Inhibitor of Apoptosis Proteins/metabolism , Neural Stem Cells/cytology , Repressor Proteins/metabolism , Wnt Proteins/metabolism , Aging/physiology , Animals , Astrocytes/cytology , Astrocytes/metabolism , Cell Differentiation/physiology , Cell Growth Processes/physiology , Hippocampus/metabolism , Humans , Mice , Neural Stem Cells/metabolism , Signal Transduction , Survivin , Wnt Signaling Pathway
12.
Nat Biotechnol ; 29(9): 824-8, 2011 Aug 10.
Article in English | MEDLINE | ID: mdl-21832997

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a fatal motor neuron disease, with astrocytes implicated as contributing substantially to motor neuron death in familial (F)ALS. However, the proposed role of astrocytes in the pathology of ALS derives in part from rodent models of FALS based upon dominant mutations within the superoxide dismutase 1 (SOD1) gene, which account for <2% of all ALS cases. Their role in sporadic (S)ALS, which affects >90% of ALS patients, remains to be established. Using astrocytes generated from postmortem tissue from both FALS and SALS patients, we show that astrocytes derived from both patient groups are similarly toxic to motor neurons. We also demonstrate that SOD1 is a viable target for SALS, as its knockdown significantly attenuates astrocyte-mediated toxicity toward motor neurons. Our data highlight astrocytes as a non-cell autonomous component in SALS and provide an in vitro model system to investigate common disease mechanisms and evaluate potential therapies for SALS and FALS.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Astrocytes/pathology , Motor Neurons/pathology , Amyotrophic Lateral Sclerosis/pathology , Animals , Astrocytes/metabolism , Biomarkers , Cell Differentiation , Cell Line , Coculture Techniques , Disease Models, Animal , Embryonic Stem Cells/cytology , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Gene Expression Regulation , Humans , Immunohistochemistry , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice , Motor Neurons/cytology , Motor Neurons/drug effects , Mutation , Sequence Analysis, DNA , Signal Transduction , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Superoxide Dismutase-1
13.
Mol Ther ; 19(11): 1971-80, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21811247

ABSTRACT

Adeno-associated virus type 9 (AAV9) is a powerful tool for delivering genes throughout the central nervous system (CNS) following intravenous injection. Preclinical results in pediatric models of spinal muscular atrophy (SMA) and lysosomal storage disorders provide a compelling case for advancing AAV9 to the clinic. An important translational step is to demonstrate efficient CNS targeting in large animals at various ages. In the present study, we tested systemically injected AAV9 in cynomolgus macaques, administered at birth through 3 years of age for targeting CNS and peripheral tissues. We show that AAV9 was efficient at crossing the blood-brain barrier (BBB) at all time points investigated. Transgene expression was detected primarily in glial cells throughout the brain, dorsal root ganglia neurons and motor neurons within the spinal cord, providing confidence for translation to SMA patients. Systemic injection also efficiently targeted skeletal muscle and peripheral organs. To specifically target the CNS, we explored AAV9 delivery to cerebrospinal fluid (CSF). CSF injection efficiently targeted motor neurons, and restricted gene expression to the CNS, providing an alternate delivery route and potentially lower manufacturing requirements for older, larger patients. Our findings support the use of AAV9 for gene transfer to the CNS for disorders in pediatric populations.


Subject(s)
Gene Transfer Techniques , Genetic Therapy , Muscular Atrophy, Spinal/therapy , Animals , Brain/metabolism , Dependovirus/genetics , Gene Expression Regulation , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , HEK293 Cells , Humans , Injections, Epidural , Injections, Intra-Arterial , Macaca , Male , Motor Neurons/metabolism , Muscle, Skeletal/metabolism , Muscular Atrophy, Spinal/genetics , Neuroglia/metabolism , Spinal Cord/metabolism , Swine , Time Factors , Transduction, Genetic , Transgenes/genetics
14.
Hum Mol Genet ; 19(20): 3895-905, 2010 Oct 15.
Article in English | MEDLINE | ID: mdl-20639395

ABSTRACT

Proximal spinal muscular atrophy (SMA) is a debilitating neurological disease marked by isolated lower motor neuron death and subsequent atrophy of skeletal muscle. Historically, SMA pathology was thought to be limited to lower motor neurons and the skeletal muscles they control, yet there are several reports describing the coincidence of cardiovascular abnormalities in SMA patients. As new therapies for SMA emerge, it is necessary to determine whether these non-neuromuscular systems need to be targeted. Therefore, we have characterized left ventricular (LV) function of SMA mice (SMN2+/+; SMNΔ7+/+; Smn-/-) and compared it with that of their unaffected littermates at 7 and 14 days of age. Anatomical and physiological measurements made by electrocardiogram and echocardiography show that affected mouse pups have a dramatic decrease in cardiac function. At 14 days of age, SMA mice have bradycardia and develop a marked dilated cardiomyopathy with a concomitant decrease in contractility. Signs of decreased cardiac function are also apparent as early as 7 days of age in SMA animals. Delivery of a survival motor neuron-1 transgene using a self-complementary adeno-associated virus serotype 9 abolished the symptom of bradycardia and significantly decreased the severity of the heart defect. We conclude that severe SMA animals have compromised cardiac function resulting at least partially from early bradycardia, which is likely attributable to aberrant autonomic signaling. Further cardiographic studies of human SMA patients are needed to clarify the clinical relevance of these findings from this SMA mouse.


Subject(s)
Bradycardia , Dependovirus/genetics , Gene Transfer Techniques , Heart Failure/physiopathology , Muscular Atrophy, Spinal/physiopathology , Survival of Motor Neuron 1 Protein/genetics , Animals , Bradycardia/genetics , Bradycardia/physiopathology , Bradycardia/therapy , Cardiomyopathy, Dilated/pathology , Cardiomyopathy, Dilated/physiopathology , Disease Models, Animal , Echocardiography , Electrocardiography , Genetic Therapy , Heart Failure/pathology , Heart Failure/therapy , Mice , Mice, Transgenic , Motor Neurons/metabolism , Muscular Atrophy, Spinal/complications , Myocardial Contraction , Nerve Tissue Proteins , SMN Complex Proteins , Ventricular Function, Left
15.
Nat Biotechnol ; 28(3): 271-4, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20190738

ABSTRACT

Spinal muscular atrophy (SMA), the most common autosomal recessive neurodegenerative disease affecting children, results in impaired motor neuron function. Despite knowledge of the pathogenic role of decreased survival motor neuron (SMN) protein levels, efforts to increase SMN have not resulted in a treatment for patients. We recently demonstrated that self-complementary adeno-associated virus 9 (scAAV9) can infect approximately 60% of motor neurons when injected intravenously into neonatal mice. Here we use scAAV9-mediated postnatal day 1 vascular gene delivery to replace SMN in SMA pups and rescue motor function, neuromuscular physiology and life span. Treatment on postnatal day 5 results in partial correction, whereas postnatal day 10 treatment has little effect, suggesting a developmental period in which scAAV9 therapy has maximal benefit. Notably, we also show extensive scAAV9-mediated motor neuron transduction after injection into a newborn cynomolgus macaque. This demonstration that scAAV9 traverses the blood-brain barrier in a nonhuman primate emphasizes the clinical potential of scAAV9 gene therapy for SMA.


Subject(s)
Gene Transfer Techniques , Motor Neurons/metabolism , Muscular Atrophy, Spinal/therapy , Survival of Motor Neuron 1 Protein/biosynthesis , Survival of Motor Neuron 1 Protein/genetics , Animals , Animals, Newborn , Dependovirus/genetics , Disease Models, Animal , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Kaplan-Meier Estimate , Macaca fascicularis , Male , Mice , Mice, Transgenic , Microscopy, Fluorescence , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/metabolism , Phenotype , Survival of Motor Neuron 1 Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...