Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
1.
Hum Mol Genet ; 32(3): 417-430, 2023 01 13.
Article in English | MEDLINE | ID: mdl-35997776

ABSTRACT

Mucopolysaccharidosis type IIIB (MPS IIIB) is an autosomal recessive lysosomal storage disease caused by mutations in the gene that encodes the protein N-acetyl-glucosaminidase (NAGLU). Defective NAGLU activity results in aberrant retention of heparan sulfate within lysosomes leading to progressive central nervous system (CNS) degeneration. Intravenous treatment options are limited by the need to overcome the blood-brain barrier and gain successful entry into the CNS. Additionally, we have demonstrated that AAV8 provides a broader transduction area in the MPS IIIB mouse brain compared with AAV5, 9 or rh10. A triple-capsid mutant (tcm) modification of AAV8 further enhanced GFP reporter expression and distribution. Using the MPS IIIB mouse model, we performed a study using either intracranial six site or intracisterna magna injection of AAVtcm8-codon-optimized (co)-NAGLU using untreated MPS IIIB mice as controls to assess disease correction. Disease correction was evaluated based on enzyme activity, heparan sulfate storage levels, CNS lysosomal signal intensity, coordination, activity level, hearing and survival. Both histologic and enzymatic assessments show that each injection method results in supranormal levels of NAGLU expression in the brain. In this study, we have shown correction of lifespan and auditory deficits, increased CNS NAGLU activity and reduced lysosomal storage levels of heparan sulfate following AAVtcm8-coNAGLU administration and partial correction of NAGLU activity in several peripheral organs in the murine model of MPS IIIB.


Subject(s)
Mucopolysaccharidosis III , Animals , Mice , Mucopolysaccharidosis III/genetics , Mucopolysaccharidosis III/therapy , Mucopolysaccharidosis III/metabolism , Capsid/metabolism , Acetylglucosaminidase/genetics , Acetylglucosaminidase/metabolism , Heparitin Sulfate/metabolism
2.
Mol Ther Methods Clin Dev ; 15: 194-203, 2019 Dec 13.
Article in English | MEDLINE | ID: mdl-31660421

ABSTRACT

Pompe disease is caused by mutations in the gene encoding the lysosomal glycogen-metabolizing enzyme, acid-alpha glucosidase (GAA). Tongue myofibers and hypoglossal motoneurons appear to be particularly susceptible in Pompe disease. Here we used intramuscular delivery of adeno-associated virus serotype 9 (AAV9) for targeted delivery of an enhanced form of GAA to tongue myofibers and motoneurons in 6-month-old Pompe (Gaa -/- ) mice. We hypothesized that addition of a glycosylation-independent lysosomal targeting tag to the protein would result in enhanced expression in tongue (hypoglossal) motoneurons when compared to the untagged GAA. Mice received an injection into the base of the tongue with AAV9 encoding either the tagged or untagged enzyme; tissues were harvested 4 months later. Both AAV9 constructs effectively drove GAA expression in lingual myofibers and hypoglossal motoneurons. However, mice treated with the AAV9 construct encoding the modified GAA enzyme had a >200% increase in the number of GAA-positive motoneurons as compared to the untagged GAA (p < 0.008). Our results confirm that tongue delivery of AAV9-encoding GAA can effectively target tongue myofibers and associated motoneurons in Pompe mice and indicate that the effectiveness of this approach can be improved by addition of the glycosylation-independent lysosomal targeting tag.

3.
Ann Transl Med ; 7(13): 290, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31392202

ABSTRACT

Pompe disease is a neuromuscular disease caused by a deficiency of the lysosomal enzyme acid alpha-glucosidase leading to lysosomal and cytoplasmic glycogen accumulation in neurons and striated muscle. In the decade since availability of first-generation enzyme replacement therapy (ERT) a better understanding of the clinical spectrum of disease has emerged. The most severe form of early onset disease is typically identified with symptoms in the first year of life, known as infantile-onset Pompe disease (IOPD). Infants are described at floppy babies with cardiac hypertrophy in the first few months of life. A milder form with late onset (LOPD) of symptoms is mostly free of cardiac involvement with slower rate of progression. Glycogen accumulation in the CNS and skeletal muscle is observed in both IOPD and LOPD. In both circumstances, multi-system disease (principally motoneuron and myopathy) leads to progressive weakness with associated respiratory and feeding difficulty. In IOPD the untreated natural history leads to cardiorespiratory failure and death in the first year of life. In the current era of ERT clinical outcomes are improved, yet, many patients have an incomplete response and a substantial unmet need remains. Since the neurological manifestations of the disease are not amenable to peripheral enzyme replacement, we set out to better understand the pathophysiology and potential for treatment of disease manifestations using adeno-associated virus (AAV)-mediated gene transfer, with the first clinical gene therapy studies initiated by our group in 2006. This review focuses on the preclinical studies and clinical study findings which are pertinent to the development of a comprehensive gene therapy strategy for both IOPD and LOPD. Given the advent of newborn screening, a significant focus of our recent work has been to establish the basis for repeat administration of AAV vectors to enhance neuromuscular therapeutic efficacy over the life span.

4.
Curr Gene Ther ; 19(3): 197-207, 2019.
Article in English | MEDLINE | ID: mdl-31223086

ABSTRACT

BACKGROUND: Pompe disease is a fatal neuromuscular disorder caused by a deficiency in acid α-glucosidase, an enzyme responsible for glycogen degradation in the lysosome. Currently, the only approved treatment for Pompe disease is enzyme replacement therapy (ERT), which increases patient survival, but does not fully correct the skeletal muscle pathology. Skeletal muscle pathology is not corrected with ERT because low cation-independent mannose-6-phosphate receptor abundance and autophagic accumulation inhibits the enzyme from reaching the lysosome. Thus, a therapy that more efficiently targets skeletal muscle pathology, such as adeno-associated virus (AAV), is needed for Pompe disease. OBJECTIVE: The goal of this project was to deliver a rAAV9-coGAA vector driven by a tissue restrictive promoter will efficiently transduce skeletal muscle and correct autophagic accumulation. METHODS: Thus, rAAV9-coGAA was intravenously delivered at three doses to 12-week old Gaa-/- mice. 1 month after injection, skeletal muscles were biochemically and histologically analyzed for autophagy-related markers. RESULTS: At the highest dose, GAA enzyme activity and vacuolization scores achieved therapeutic levels. In addition, resolution of autophagosome (AP) accumulation was seen by immunofluorescence and western blot analysis of autophagy-related proteins. Finally, mice treated at birth demonstrated persistence of GAA expression and resolution of lysosomes and APs compared to those treated at 3 months. CONCLUSION: In conclusion, a single systemic injection of rAAV9-coGAA ameliorates vacuolar accumulation and prevents autophagic dysregulation.


Subject(s)
Autophagy , Dependovirus/genetics , Genetic Therapy , Genetic Vectors/administration & dosage , Glycogen Storage Disease Type II/therapy , Muscle, Skeletal/physiology , alpha-Glucosidases/physiology , Animals , Disease Models, Animal , Enzyme Replacement Therapy/methods , Female , Glycogen Storage Disease Type II/enzymology , Glycogen Storage Disease Type II/genetics , Glycogen Storage Disease Type II/pathology , Lysosomes , Male , Mice , Mice, Knockout
5.
ACS Chem Neurosci ; 10(6): 2890-2902, 2019 06 19.
Article in English | MEDLINE | ID: mdl-31017387

ABSTRACT

Hereditary demyelinating neuropathies linked to peripheral myelin protein 22 (PMP22) involve the disruption of normal protein trafficking and are therefore relevant targets for chaperone therapy. Using a small molecule HSP90 inhibitor, EC137, in cell culture models, we previously validated the chaperone pathway as a viable target for therapy development. Here, we tested five commercially available inhibitors of HSP90 and identified BIIB021 and AUY922 to support Schwann cell viability and enhance chaperone expression. AUY922 showed higher efficacy, compared to BIIB021, in enhancing myelin synthesis in dorsal root ganglion explant cultures from neuropathic mice. For in vivo testing, we randomly assigned 2-3 month old C22 and 6 week old Trembler J (TrJ) mice to receive two weekly injections of either vehicle or AUY922 (2 mg/kg). By the intraperitoneal (i.p.) route, the drug was well-tolerated by all mice over the 5 month long study, without influence on body weight or general grooming behavior. AUY922 improved the maintenance of myelinated nerves of both neuropathic models and attenuated the decline in rotarod performance and peak muscle force production in C22 mice. These studies highlight the significance of proteostasis in neuromuscular function and further validate the HSP90 pathway as a therapeutic target for hereditary neuropathies.


Subject(s)
Charcot-Marie-Tooth Disease/pathology , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Isoxazoles/pharmacology , Nerve Fibers, Myelinated/drug effects , Resorcinols/pharmacology , Animals , Axons/drug effects , Axons/pathology , Mice , Myelin Sheath/drug effects , Myelin Sheath/pathology , Nerve Fibers, Myelinated/pathology
6.
Muscle Nerve ; 57(4): 664-671, 2018 04.
Article in English | MEDLINE | ID: mdl-29023846

ABSTRACT

INTRODUCTION: Patients with hereditary peripheral neuropathies exhibit characteristic deformities of the hands and feet and have difficulty ambulating. To examine to what extent neuropathic animals recapitulate these deficits, we studied trembler J (TrJ) mice, which model early-onset demyelinating neuropathy. METHODS: A cohort of 4-month-old female wild type and neuropathic mice were evaluated for locomotor measurements, neuromuscular function, and skeletal muscle proteolysis and morphometry. RESULTS: Utilizing the DigiGait imaging system, we identified pronounced alterations in forepaw and hindpaw angles and a decrease in hindpaw area on the treadmill in neuropathic rodents. Torque production by the tibialis anterior (TA) muscle was significantly weakened and was paralleled by a decrease in myofiber cross-sectional area and an increase in muscle tissue proteolysis. DISCUSSION: Our findings in TrJ mice reflect the phenotypic presentation of the human neuropathy in which patients exhibit weakness of the TA muscle resulting in foot drop and locomotor abnormalities. Muscle Nerve 57: 664-671, 2018.


Subject(s)
Charcot-Marie-Tooth Disease/physiopathology , Locomotion/physiology , Muscle, Skeletal/physiopathology , Animals , Charcot-Marie-Tooth Disease/genetics , Disease Models, Animal , Female , Gait Analysis , Hereditary Sensory and Motor Neuropathy/genetics , Hereditary Sensory and Motor Neuropathy/physiopathology , Mice , Myelin Proteins/genetics , Peripheral Nervous System Diseases/genetics , Peripheral Nervous System Diseases/physiopathology , Phenotype , Torque
8.
Mol Neurobiol ; 55(6): 5299-5309, 2018 Jun.
Article in English | MEDLINE | ID: mdl-28895054

ABSTRACT

In Pompe disease, deficient lysosomal acid α-glucosidase (GAA) activity causes glycogen accumulation in the muscles, which leads to weakness, cardiomyopathy, and respiratory failure. Although glycogen accumulation also occurs in the nervous system, the burden of neurological deficits in Pompe disease remains obscure. In this study, a neuron-specific gene therapy was administered to Pompe mice through intracerebroventricular injection of a viral vector carrying a neuron-specific promoter. The results revealed that gene therapy increased GAA activity and decreased glycogen content in the brain and spinal cord but not in the muscles of Pompe mice. Gene therapy only slightly increased the muscle strength of Pompe mice but substantially improved their performance on the rotarod, a test measuring motor coordination. Gene therapy also decreased astrogliosis and increased myelination in the brain and spinal cord of Pompe mice. Therefore, a neuron-specific treatment improved the motor coordination of Pompe mice by lowering glycogen accumulation, decreasing astrogliosis, and increasing myelination. These findings indicate that neurological deficits are responsible for a significant burden in Pompe disease.


Subject(s)
Genetic Therapy , Glycogen Storage Disease Type II/physiopathology , Glycogen Storage Disease Type II/therapy , Motor Activity , Neurons/metabolism , Animals , Brain/pathology , Gliosis/metabolism , Gliosis/pathology , Glycogen/metabolism , Glycogen Storage Disease Type II/genetics , Mice , Muscle Strength , Myelin Sheath/metabolism , Respiration , Rotarod Performance Test , Spinal Cord/pathology , Tissue Distribution , alpha-Glucosidases/genetics , alpha-Glucosidases/therapeutic use
9.
Mol Ther Methods Clin Dev ; 7: 42-49, 2017 Dec 15.
Article in English | MEDLINE | ID: mdl-29018835

ABSTRACT

The development of therapeutic clinical trials for glycogen storage disorders, including Pompe disease, has called for non-invasive and objective biomarkers. Glycogen accumulation can be measured in vivo with 13C MRS. However, clinical implementation remains challenging due to low signal-to-noise. On the other hand, the buildup of glycolytic intermediates may be detected with 31P MRS. We sought to identify new biomarkers of disease progression in muscle using 13C/31P MRS and 1H HR-MAS in a mouse model of Pompe disease (Gaa-/-). We evaluated the sensitivity of these MR biomarkers in vivo after treatment using an adeno-associated virus vector 2/9 encoding hGAA driven by the desmin promotor. 31P MRS showed significantly elevated phosphomonoesters (PMEs) in Gaa-/- compared to control at 2 (0.06 ± 0.02 versus 0.03 ± 0.01; p = 0.003), 6, 12, and 18 months of age. Correlative 1H HR-MAS measures in intact gastrocnemius muscles revealed high glucose-6-phosphate (G-6-P). After intramuscular AAV injections, glycogen, PME, and G-6-P were decreased within normal range. The changes in PME levels likely partly resulted from changes in G-6-P, one of the overlapping phosphomonoesters in the 31P MR spectra in vivo. Because 31P MRS is inherently more sensitive than 13C MRS, PME levels have greater potential as a clinical biomarker and should be considered as a complementary approach for future studies in Pompe patients.

10.
Mol Ther Methods Clin Dev ; 3: 16031, 2016.
Article in English | MEDLINE | ID: mdl-27222839

ABSTRACT

Recombinant adeno-associated vectors based on serotype 9 (rAAV9) have demonstrated highly effective gene transfer in multiple animal models of muscular dystrophies and other neurological indications. Current limitations in vector production and purification have hampered widespread implementation of clinical candidate vectors, particularly when systemic administration is considered. In this study, we describe a complete herpes simplex virus (HSV)-based production and purification process capable of generating greater than 1 × 10(14) rAAV9 vector genomes per 10-layer CellSTACK of HEK 293 producer cells, or greater than 1 × 10(5) vector genome per cell, in a final, fully purified product. This represents a 5- to 10-fold increase over transfection-based methods. In addition, rAAV vectors produced by this method demonstrated improved biological characteristics when compared to transfection-based production, including increased infectivity as shown by higher transducing unit-to-vector genome ratios and decreased total capsid protein amounts, shown by lower empty-to-full ratios. Together, this data establishes a significant improvement in both rAAV9 yields and vector quality. Further, the method can be readily adapted to large-scale good laboratory practice (GLP) and good manufacturing practice (GMP) production of rAAV9 vectors to enable preclinical and clinical studies and provide a platform to build on toward late-phases and commercial production.

11.
Respir Physiol Neurobiol ; 227: 48-55, 2016 Jun 15.
Article in English | MEDLINE | ID: mdl-26921786

ABSTRACT

Respiratory and/or lingual dysfunction are among the first motor symptoms in Pompe disease, a disorder resulting from absence or dysfunction of the lysosomal enzyme acid α-glucosidase (GAA). Here, we histologically evaluated the medulla, cervical and thoracic spinal cords in 6 weeks old asymptomatic Pompe (Gaa(-/-)) mice to determine if neuropathology in respiratory motor regions has an early onset. Periodic acid-Schiff (PAS) staining indicated glycogen accumulation was exclusively occurring in Gaa(-/-) hypoglossal, mid-cervical and upper thoracic motoneurons. Markers of DNA damage (Tunel) and ongoing apoptosis (Cleaved Caspase 3) did not co-localize with PAS staining, but were prominent in a medullary region which included the nucleus tractus solitarius, and also in the thoracic spinal dorsal horn. We conclude that respiratory-related motoneurons are particularly susceptible to GAA deficiency and that neuronal glycogen accumulation and neurodegeneration may occur independently in early stage disease. The data support early therapeutic intervention in Pompe disease.


Subject(s)
Glycogen Storage Disease Type II/pathology , Medulla Oblongata/pathology , Motor Neurons/pathology , Spinal Cord/pathology , Animals , Apoptosis , Calcium-Binding Proteins/metabolism , Caspase 3/metabolism , Cervical Vertebrae , Cohort Studies , DNA Damage , Disease Models, Animal , Glial Fibrillary Acidic Protein/metabolism , Glycogen/metabolism , Glycogen Storage Disease Type II/metabolism , Medulla Oblongata/metabolism , Mice, 129 Strain , Mice, Knockout , Microfilament Proteins/metabolism , Motor Neurons/metabolism , Neuroimmunomodulation , Spinal Cord/metabolism , Thoracic Vertebrae
12.
Hum Gene Ther ; 27(1): 43-59, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26603344

ABSTRACT

Pompe disease is a progressive neuromuscular disorder caused by lysosomal accumulation of glycogen from a deficiency in acid alpha-glucosidase (GAA). Replacement of the missing enzyme is available by repeated protein infusions; however, efficacy is limited by immune response and inability to restore enzymatic function in the central nervous system. An alternative therapeutic option is adeno-associated virus (AAV)-mediated gene therapy, which results in widespread gene transfer and prolonged transgene expression. Both enzyme replacement therapy (ERT) and gene therapy can elicit anti-GAA immune reactions that dampen their effectiveness and pose life-threatening risks to patient safety. To modulate the immune responses related to gene therapy, we show that a human codon-optimized GAA (coGAA) driven by a liver-specific promoter (LSP) using AAV9 is capable of promoting immune tolerance in a Gaa(-/-) mouse model. Copackaging AAV9-LSP-coGAA with the tissue-restricted desmin promoter (AAV9-DES-coGAA) demonstrates the necessary cell autonomous expression in cardiac muscle, skeletal muscle, peripheral nerve, and the spinal cord. Simultaneous high-level expression in liver led to the expansion of GAA-specific regulatory T-cells (Tregs) and induction of immune tolerance. Transfer of Tregs into naïve recipients prevented pathogenic allergic reactions after repeated ERT challenges. Copackaged AAV9 also attenuated preexisting humoral and cellular immune responses, which enhanced the biochemical correction. Our data present a therapeutic design in which simultaneous administration of two copackaged AAV constructs may provide therapeutic benefit and resolve immune reactions in the treatment of multisystem disorders.


Subject(s)
Genetic Therapy , Glucan 1,4-alpha-Glucosidase/genetics , Glycogen Storage Disease Type II/genetics , Immune Tolerance/genetics , Animals , Dependovirus/genetics , Dependovirus/immunology , Glucan 1,4-alpha-Glucosidase/deficiency , Glycogen Storage Disease Type II/immunology , Glycogen Storage Disease Type II/therapy , Humans , Mice , Mice, Knockout , Promoter Regions, Genetic , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism
13.
Mol Ther Methods Clin Dev ; 2: 15007, 2015.
Article in English | MEDLINE | ID: mdl-26029718

ABSTRACT

Pompe disease is an autosomal recessive genetic disorder characterized by a deficiency of the enzyme responsible for degradation of lysosomal glycogen (acid α-glucosidase (GAA)). Cardiac dysfunction and respiratory muscle weakness are primary features of this disorder. To attenuate the progressive and rapid accumulation of glycogen resulting in cardiorespiratory dysfunction, adult Gaa (-/-) mice were administered a single systemic injection of rAAV2/9-DES-hGAA (AAV9-DES) or bimonthly injections of recombinant human GAA (enzyme replacement therapy (ERT)). Assessment of cardiac function and morphology was measured 1 and 3 months after initiation of treatment while whole-body plethysmography and diaphragmatic contractile function was evaluated at 3 months post-treatment in all groups. Gaa (-/-) animals receiving either AAV9-DES or ERT demonstrated a significant improvement in cardiac function and diaphragmatic contractile function as compared to control animals. AAV9-DES treatment resulted in a significant reduction in cardiac dimension (end diastolic left ventricular mass/gram wet weight; EDMc) at 3 months postinjection. Neither AAV nor ERT therapy altered minute ventilation during quiet breathing (eupnea). However, breathing frequency and expiratory time were significantly improved in AAV9-DES animals. These results indicate systemic delivery of either strategy improves cardiac function but AAV9-DES alone improves respiratory parameters at 3 months post-treatment in a murine model of Pompe disease.

14.
Ann Neurol ; 78(2): 222-34, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25925726

ABSTRACT

OBJECTIVE: We have recently reported on the pathology of the neuromuscular junction (NMJ) in Pompe disease, reflecting disruption of neuronal and muscle homeostasis as a result of glycogen accumulation. The aim of this study was to examine how the alteration of NMJ physiology contributes to Pompe disease pathology; we performed molecular, physiological, and histochemical analyses of NMJ-related measures of the tibialis anterior muscles of young-, mid-, and late-stage alpha-glucosidase (GAA)-deficient mice. METHODS: We performed intramuscular injection of an adeno-associated virus (AAV)9 vector expressing GAA (AAV9-hGAA) into the tibialis anterior muscle of Gaa(-/-) mice at early, mid, and severe pathological time points. We analyzed expression of NMJ-related genes, in situ muscle force production, and clearance of glycogen in conjunction with histological assessment of the NMJ. RESULTS: Our data demonstrate that AAV9-hGAA is able to replace GAA to the affected tissue and modify AChR mRNA expression, muscle force production, motor endplate area, and innervation status. Importantly, the degree of restoration for these outcomes is limited by severity of disease. Early restoration of GAA activity was most effective, whereas late correction of GAA expression was not effective in modifying parameters reflecting NMJ structure and function nor in force restoration despite resolution of glycogen storage in muscle. INTERPRETATION: Our data provide new mechanistic insight into the pathology of Pompe disease and suggest that early systemic correction to both neural and muscle tissues may be essential for successful correction of neuromuscular function in Pompe disease. Ann Neurol 2015;78:222-234.


Subject(s)
Genetic Therapy , Glycogen Storage Disease Type II/genetics , Muscle Strength/physiology , Muscle, Skeletal/metabolism , Neuromuscular Junction/metabolism , RNA, Messenger/metabolism , Receptors, Cholinergic/genetics , alpha-Glucosidases/genetics , Animals , Dependovirus , Disease Models, Animal , Genetic Vectors , Glycogen/metabolism , Glycogen Storage Disease Type II/metabolism , Glycogen Storage Disease Type II/physiopathology , Hindlimb , Injections, Intramuscular , Isometric Contraction , Mice , Mice, Knockout , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Neuromuscular Junction/pathology , Neuromuscular Junction/physiopathology , Receptors, Cholinergic/metabolism , Time Factors
15.
J Inherit Metab Dis ; 38(5): 915-22, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25860817

ABSTRACT

Barth syndrome (BTHS) is an X-linked metabolic disorder that causes cardiomyopathy in infancy and is linked to mutations within the Tafazzin (TAZ) gene. The first mouse model, a TAZ knockdown model (TAZKD), has been generated to further understand the bioenergetics leading to cardiomyopathy. However, the TAZKD model does not show early signs of cardiomyopathy, and cardiac pathophysiology has not been documented until 7-8 months of age. Here we sought to determine the impact of endurance training on the cardiac and skeletal muscle phenotype in young TAZKD mice. TAZKD exercise trained (TAZKD-ET) and control exercise trained (CON-ET) mice underwent a 35-day swimming protocol. Non-trained aged matched TAZKD and CON mice were used as controls. At the end of the protocol, cardiac MRI was used to assess cardiac parameters. Cardiac MRI showed that training resulted in cardiac hypertrophy within both groups and did not result in a decline of ejection fraction. TAZKD mice exhibited a decrease in respiratory complex I, III, and IV enzymatic activity in cardiac tissue compared to control mice; however, training led to an increase in complex III activity in TAZKD-ET mice resulting in similar levels to those of CON-ET mice. (31)P magnetic resonance spectroscopy of the gastrocnemius showed a significantly lowered pH in TAZKD-ET mice post electrical-stimulation compared to CON-ET mice. Endurance training does not accelerate cardiac dysfunction in young TAZKD mice, but results in beneficial physiological effects. Furthermore, our results suggest that a significant drop in intracellular pH levels may contribute to oxidative phosphorylation defects during exercise.


Subject(s)
Barth Syndrome/pathology , Barth Syndrome/therapy , Disease Models, Animal , Electron Transport Complex III/deficiency , Physical Conditioning, Animal/physiology , Physical Endurance , Transcription Factors/genetics , Acyltransferases , Animals , Barth Syndrome/genetics , Electron Transport Complex III/genetics , Exercise/physiology , Exercise Tolerance/genetics , Humans , Mice , Mice, Knockout , Physical Endurance/genetics , Physical Endurance/physiology , Reactive Oxygen Species/metabolism
16.
Am J Respir Cell Mol Biol ; 53(3): 326-35, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25569118

ABSTRACT

Pompe disease results from a mutation in the acid α-glucosidase gene leading to lysosomal glycogen accumulation. Respiratory insufficiency is common, and the current U.S. Food and Drug Administration-approved treatment, enzyme replacement, has limited effectiveness. Ampakines are drugs that enhance α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor responses and can increase respiratory motor drive. Recent work indicates that respiratory motor drive can be blunted in Pompe disease, and thus pharmacologic stimulation of breathing may be beneficial. Using a murine Pompe model with the most severe clinical genotype (the Gaa(-/-) mouse), our primary objective was to test the hypothesis that ampakines can stimulate respiratory motor output and increase ventilation. Our second objective was to confirm that neuropathology was present in Pompe mouse medullary respiratory control neurons. The impact of ampakine CX717 on breathing was determined via phrenic and hypoglossal nerve recordings in anesthetized mice and whole-body plethysmography in unanesthetized mice. The medulla was examined using standard histological methods coupled with immunochemical markers of respiratory control neurons. Ampakine CX717 robustly increased phrenic and hypoglossal inspiratory bursting and reduced respiratory cycle variability in anesthetized Pompe mice, and it increased inspiratory tidal volume in unanesthetized Pompe mice. CX717 did not significantly alter these variables in wild-type mice. Medullary respiratory neurons showed extensive histopathology in Pompe mice. Ampakines stimulate respiratory neuromotor output and ventilation in Pompe mice, and therefore they have potential as an adjunctive therapy in Pompe disease.


Subject(s)
Glycogen Storage Disease Type II/drug therapy , Isoxazoles/pharmacology , Respiration/drug effects , Respiratory System Agents/pharmacology , Animals , Brain Stem/pathology , Drug Evaluation, Preclinical , Glycogen Storage Disease Type II/physiopathology , Isoxazoles/therapeutic use , Mice, 129 Strain , Mice, Knockout , Motor Activity/drug effects , Phrenic Nerve/drug effects , Phrenic Nerve/physiopathology , Respiratory System Agents/therapeutic use
17.
Hum Mol Genet ; 24(3): 625-36, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25217571

ABSTRACT

Pompe disease is a systemic metabolic disorder characterized by lack of acid-alpha glucosidase (GAA) resulting in ubiquitous lysosomal glycogen accumulation. Respiratory and ambulatory dysfunction are prominent features in patients with Pompe yet the mechanism defining the development of muscle weakness is currently unclear. Transgenic animal models of Pompe disease mirroring the patient phenotype have been invaluable in mechanistic and therapeutic study. Here, we demonstrate significant pathological alterations at neuromuscular junctions (NMJs) of the diaphragm and tibialis anterior muscle as prominent features of disease pathology in Gaa knockout mice. Postsynaptic defects including increased motor endplate area and fragmentation were readily observed in Gaa(-/-) but not wild-type mice. Presynaptic neuropathic changes were also evident, as demonstrated by significant reduction in the levels of neurofilament proteins, and alterations in axonal fiber diameter and myelin thickness within the sciatic and phrenic nerves. Our data suggest the loss of NMJ integrity is a primary contributor to the decline in respiratory and ambulatory function in Pompe and arises from both pre- and postsynaptic pathology. These observations highlight the importance of systemic phenotype correction, specifically restoration of GAA to skeletal muscle and the nervous system for treatment of Pompe disease.


Subject(s)
Glycogen Storage Disease Type II/pathology , Membrane Glycoproteins/metabolism , Muscle, Skeletal/pathology , Neuromuscular Junction/pathology , Phrenic Nerve/pathology , Animals , Diaphragm/metabolism , Diaphragm/pathology , Disease Models, Animal , Glycogen Storage Disease Type II/genetics , Glycogen Storage Disease Type II/metabolism , Humans , Membrane Glycoproteins/genetics , Mice , Mice, Knockout , Muscle, Skeletal/metabolism , Neuromuscular Junction/metabolism , Phrenic Nerve/metabolism , Tibia/metabolism , Tibia/pathology
18.
Muscle Nerve ; 51(6): 877-83, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25186912

ABSTRACT

INTRODUCTION: Pompe disease is a progressive disease that affects skeletal muscles and leads to loss of ambulation. We investigated the activation of the tibialis anterior (TA) in late-onset Pompe disease (LOPD) individuals during maximal voluntary contraction (MVC) and evoked involuntary responses. METHODS: Four LOPD patients and matched control subjects performed MVC of the TA using dorsiflexion and TA evoked responses. Activation of the TA was recorded with surface electromyography. RESULTS: The Pompe patients exhibited greater power at frequencies below 60 Hz and reduced power above 100 Hz. They also exhibited a reduced increase in M-wave and prolonged M-wave latency and duration in response to stimulation. CONCLUSIONS: These results provide evidence that LOPD individuals have an altered activation pattern of the TA during maximal contractions. The observed activation pattern may reflect impairments in voluntary command, neuromuscular junction pathology, or compensatory drive due to a reduced number of functional motoneurons.


Subject(s)
Glycogen Storage Disease Type II/complications , Isometric Contraction/physiology , Movement Disorders/etiology , Muscle, Skeletal/physiopathology , Adolescent , Adult , Electric Stimulation , Evoked Potentials, Motor/physiology , Female , Humans , Male , Young Adult
19.
Neurobiol Dis ; 70: 224-36, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25014022

ABSTRACT

Charcot--Marie-Tooth disease type 1A (CMT1A) is a hereditary peripheral neuropathy characterized by progressive demyelination and distal muscle weakness. Abnormal expression of peripheral myelin protein 22 (PMP22) has been linked to CMT1A and is modeled by Trembler J (TrJ) mice, which carry the same leucine to proline substitution in PMP22 as affected pedigrees. Pharmacologic modulation of autophagy by rapamycin in neuron-Schwann cell explant cultures from neuropathic mice reduced PMP22 aggregate formation and improved myelination. Here we asked whether rapamycin administration by food supplementation, or intraperitoneal injection, could alleviate the neuropathic phenotype of affected mice and improve neuromuscular performance. Cohorts of male and female wild type (Wt) and TrJ mice were assigned to placebo or rapamycin treatment starting at 2 or 4months of age and tested monthly on the rotarod. While neither long-term feeding (8 or 10months) on rapamycin-enriched diet, or short-term injection (2months) of rapamycin improved locomotor performance of the neuropathic mice, both regimen benefited peripheral nerve myelination. Together, these results indicate that while treatment with rapamycin benefits the myelination capacity of neuropathic Schwann cells, this intervention does not improve neuromuscular function. The observed outcome might be the result of the differential response of nerve and skeletal muscle tissue to rapamycin.


Subject(s)
Motor Activity/drug effects , Neurons/drug effects , Neuroprotective Agents/administration & dosage , Peripheral Nervous System Diseases/drug therapy , Schwann Cells/drug effects , Sirolimus/administration & dosage , Animals , Charcot-Marie-Tooth Disease , Cohort Studies , Dietary Supplements , Female , Injections, Intraperitoneal , Male , Mice , Mice, Inbred C57BL , Motor Activity/physiology , Muscle, Skeletal/drug effects , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Mutation , Myelin Sheath/drug effects , Myelin Sheath/pathology , Myelin Sheath/physiology , Neurons/pathology , Neurons/physiology , Peripheral Nervous System Diseases/pathology , Peripheral Nervous System Diseases/physiopathology , Random Allocation , Rotarod Performance Test , Schwann Cells/pathology , Schwann Cells/physiology , Tissue Culture Techniques
20.
Mol Ther ; 22(4): 702-12, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24336173

ABSTRACT

Pompe disease is an autosomal recessive disorder caused by mutations in the acid-α glucosidase (GAA) gene. Lingual dysfunction is prominent but does not respond to conventional enzyme replacement therapy (ERT). Using Pompe (Gaa(-/-)) mice, we tested the hypothesis that intralingual delivery of viral vectors encoding GAA results in GAA expression and glycogen clearance in both tongue myofibers and hypoglossal (XII) motoneurons. An intralingual injection of an adeno-associated virus (AAV) vector encoding GAA (serotypes 1 or 9; 1 × 10(11) vector genomes, CMV promoter) was performed in 2-month-old Gaa(-/-) mice, and tissues were harvested 4 months later. Both serotypes robustly transduced tongue myofibers with histological confirmation of GAA expression (immunochemistry) and glycogen clearance (Period acid-Schiff stain). Both vectors also led to medullary transgene expression. GAA-positive motoneurons did not show the histopathologic features which are typical in Pompe disease and animal models. Intralingual injection with the AAV9 vector resulted in approximately threefold more GAA-positive XII motoneurons (P < 0.02 versus AAV1); the AAV9 group also gained more body weight over the course of the study (P < 0.05 versus AAV1 and sham). We conclude that intralingual injection of AAV1 or AAV9 drives persistent GAA expression in tongue myofibers and motoneurons, but AAV9 may more effectively target motoneurons.


Subject(s)
Genetic Therapy , Glycogen Storage Disease Type II/genetics , Glycogen Storage Disease Type II/therapy , Motor Neurons/metabolism , alpha-Glucosidases/genetics , Animals , Dependovirus/genetics , Gene Expression Regulation, Enzymologic , Gene Transfer Techniques , Glycogen , Glycogen Storage Disease Type II/pathology , Humans , Injections, Intramuscular , Mice , Motor Neurons/pathology , Muscle, Skeletal/metabolism , Myofibrils/genetics , Myofibrils/metabolism , Promoter Regions, Genetic , alpha-Glucosidases/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...