Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
Ann Neurol ; 77(3): 399-414, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25516063

ABSTRACT

OBJECTIVES: Spinal muscular atrophy (SMA) is caused by reduced levels of survival motor neuron (SMN) protein, which results in motoneuron loss. Therapeutic strategies to increase SMN levels including drug compounds, antisense oligonucleotides, and scAAV9 gene therapy have proved effective in mice. We wished to determine whether reduction of SMN in postnatal motoneurons resulted in SMA in a large animal model, whether SMA could be corrected after development of muscle weakness, and the response of clinically relevant biomarkers. METHODS: Using intrathecal delivery of scAAV9 expressing an shRNA targeting pig SMN1, SMN was knocked down in motoneurons postnatally to SMA levels. This resulted in an SMA phenotype representing the first large animal model of SMA. Restoration of SMN was performed at different time points with scAAV9 expressing human SMN (scAAV9-SMN), and electrophysiology measurements and pathology were performed. RESULTS: Knockdown of SMN in postnatal motoneurons results in overt proximal weakness, fibrillations on electromyography indicating active denervation, and reduced compound muscle action potential (CMAP) and motor unit number estimation (MUNE), as in human SMA. Neuropathology showed loss of motoneurons and motor axons. Presymptomatic delivery of scAAV9-SMN prevented SMA symptoms, indicating that all changes are SMN dependent. Delivery of scAAV9-SMN after symptom onset had a marked impact on phenotype, electrophysiological measures, and pathology. INTERPRETATION: High SMN levels are critical in postnatal motoneurons, and reduction of SMN results in an SMA phenotype that is SMN dependent. Importantly, clinically relevant biomarkers including CMAP and MUNE are responsive to SMN restoration, and abrogation of phenotype can be achieved even after symptom onset.


Subject(s)
Disease Models, Animal , Genetic Therapy/methods , Motor Neurons/metabolism , Muscular Atrophy, Spinal/therapy , SMN Complex Proteins/metabolism , Animals , Biomarkers , Dependovirus/genetics , Electromyography , Genetic Vectors/therapeutic use , Humans , Motor Neurons/pathology , Muscular Atrophy, Spinal/etiology , Muscular Atrophy, Spinal/pathology , Muscular Atrophy, Spinal/physiopathology , Phenotype , RNA, Small Interfering/therapeutic use , SMN Complex Proteins/genetics , Swine
2.
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
3.
Hum Mol Genet ; 22(24): 4929-37, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-23863459

ABSTRACT

Pharmacologic strategies have provided modest improvement in the devastating muscle-wasting disease, Duchenne muscular dystrophy (DMD). Pre-clinical gene therapy studies have shown promise in the mdx mouse model; however, studies conducted after disease onset fall short of fully correcting muscle strength or protecting against contraction-induced injury. Here we examine the treatment effect on muscle physiology in aged dystrophic mice with significant disease pathology by combining two promising therapies: micro-dystrophin gene replacement and muscle enhancement with follistatin, a potent myostatin inhibitor. Individual treatments with micro-dystrophin and follistatin demonstrated marked improvement in mdx mice but were insufficient to fully restore muscle strength and response to injury to wild-type levels. Strikingly, when combined, micro-dystrophin/follistatin treatment restored force generation and conferred resistance to contraction-induced injury in aged mdx mice. Pre-clinical studies with miniature dystrophins have failed to demonstrate full correction of the physiological defects seen in mdx mice. Importantly, the addition of a muscle enhancement strategy with delivery of follistatin in combination with micro-dystrophin gene therapy completely restored resistance to eccentric contraction-induced injury and improved force. Eccentric contraction-induced injury is a pre-clinical parameter relevant to the exercise induced injury that occurs in DMD patients, and herein, we demonstrate compelling evidence for the therapeutic potential of micro-dystrophin/follistatin combinatorial therapy.


Subject(s)
Dystrophin/genetics , Follistatin/genetics , Muscle, Skeletal/metabolism , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/metabolism , Animals , Dependovirus/genetics , Disease Models, Animal , Dystrophin/metabolism , Follistatin/metabolism , Gene Expression , Gene Transfer Techniques , Genetic Therapy , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Mice , Mice, Inbred mdx , Muscle Contraction/genetics , Muscle Strength/genetics , Muscle, Skeletal/pathology , Muscular Dystrophy, Animal , Muscular Dystrophy, Duchenne/therapy
4.
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
5.
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
6.
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
7.
Ann Clin Transl Neurol ; 1(1): 34-44, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24511555

ABSTRACT

OBJECTIVE: Preclinical therapies that restore survival motor neuron (SMN) protein levels can dramatically extend survival in spinal muscular atrophy (SMA) mouse models. Biomarkers are needed to effectively translate these promising therapies to clinical trials. Our objective was to investigate electrophysiological biomarkers of compound muscle action potential (CMAP), motor unit number estimation (MUNE) and electromyography (EMG) using an SMA mouse model. METHODS: Sciatic CMAP, MUNE, and EMG were obtained in SMNΔ7 mice at ages 3-13 days and at 21 days in mice with SMN selectively reduced in motor neurons (ChATCre ). To investigate these measures as biomarkers of treatment response, measurements were obtained in SMNΔ7 mice treated with antisense oligonucleotide (ASO) or gene therapy. RESULTS: CMAP was significantly reduced in SMNΔ7 mice at days 6-13 (p<0.01), and MUNE was reduced at days 7-13 (p<0.01). Fibrillations were present on EMG in SMNΔ7 mice but not controls (p=0.02). Similar findings were seen at 21 days in ChATCre mice. MUNE in ASO-treated SMNΔ7 mice were similar to controls at day 12 and 30. CMAP reduction persisted in ASO-treated SMNΔ7 mice at day 12 but was corrected at day 30. Similarly, CMAP and MUNE responses were corrected with gene therapy to restore SMN. INTERPRETATION: These studies confirm features of preserved neuromuscular function in the early postnatal period and subsequent motor unit loss in SMNΔ7 mice. SMN restoring therapies result in preserved MUNE and gradual repair of CMAP responses. This provides preclinical evidence for the utilization of CMAP and MUNE as biomarkers in future SMA clinical trials.

8.
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
SELECTION OF CITATIONS
SEARCH DETAIL