Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Am J Physiol Lung Cell Mol Physiol ; 325(3): L288-L298, 2023 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-37366541

RESUMEN

Pompe disease is an autosomal recessive glycogen storage disease caused by mutations in the gene that encodes acid alpha-glucosidase (GAA)-an enzyme responsible for hydrolyzing lysosomal glycogen. GAA deficiency results in systemic lysosomal glycogen accumulation and cellular disruption. Glycogen accumulation in skeletal muscles, motor neurons, and airway smooth muscle cells is known to contribute to respiratory insufficiency in Pompe disease. However, the impact of GAA deficiency on the distal alveolar type 1 and type 2 cells (AT1 and AT2) has not been evaluated. AT1 cells rely on lysosomes for cellular homeostasis so that they can maintain a thin barrier for gas exchange, whereas AT2 cells depend on lysosome-like structures (lamellar bodies) for surfactant production. Using a mouse model of Pompe disease, the Gaa-/- mouse, we investigated the consequences of GAA deficiency on AT1 and AT2 cells using histology, pulmonary function and mechanics, and transcriptional analysis. Histological analysis revealed increased accumulation of lysosomal-associated membrane protein 1 (LAMP1) in the Gaa-/- mice lungs. Furthermore, ultrastructural examination showed extensive intracytoplasmic vacuoles enlargement and lamellar body engorgement. Respiratory dysfunction was confirmed using whole body plethysmography and forced oscillometry. Finally, transcriptomic analysis demonstrated dysregulation of surfactant proteins in AT2 cells, specifically reduced levels of surfactant protein D in the Gaa-/- mice. We conclude that GAA enzyme deficiency leads to glycogen accumulation in the distal airway cells that disrupts surfactant homeostasis and contributes to respiratory impairments in Pompe disease.NEW & NOTEWORTHY This research highlights the impact of Pompe disease on distal airway cells. Prior to this work, respiratory insufficiency in Pompe disease was classically attributed to pathology in respiratory muscles and motor neurons. Using the Pompe mouse model, we note significant pathology in alveolar type 1 and 2 cells with reductions in surfactant protein D and disrupted surfactant homeostasis. These novel findings highlight the potential contributions of alveolar pathology to respiratory insufficiency in Pompe disease.


Asunto(s)
Enfermedad del Almacenamiento de Glucógeno Tipo II , Insuficiencia Respiratoria , Humanos , Enfermedad del Almacenamiento de Glucógeno Tipo II/genética , Enfermedad del Almacenamiento de Glucógeno Tipo II/patología , Proteína D Asociada a Surfactante Pulmonar/metabolismo , alfa-Glucosidasas/genética , alfa-Glucosidasas/metabolismo , Músculo Esquelético/metabolismo , Glucógeno/metabolismo
2.
J Physiol ; 600(15): 3465-3482, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35620971

RESUMEN

Duchenne muscular dystrophy (DMD) is an X-linked neuromuscular disease caused by a deficiency in dystrophin - a structural protein which stabilises muscle during contraction. Dystrophin deficiency adversely affects the respiratory system leading to sleep-disordered breathing, hypoventilation, and weakness of the expiratory and inspiratory musculature, which culminate in severe respiratory dysfunction. Muscle degeneration-associated respiratory impairment in neuromuscular disease is a result of disruptions at multiple sites of the respiratory control network, including sensory and motor pathways. As a result of this pathology, respiratory failure is a leading cause of premature death in DMD patients. Currently available treatments for DMD respiratory insufficiency attenuate respiratory symptoms without completely reversing the underlying pathophysiology. This underscores the need to develop curative therapies to improve quality of life and longevity of DMD patients. This review summarises research findings on the pathophysiology of respiratory insufficiencies in DMD disease in humans and animal models, the clinical interventions available to ameliorate symptoms, and gene-based therapeutic strategies uncovered by preclinical animal studies.


Asunto(s)
Distrofia Muscular de Duchenne , Enfermedades Neuromusculares , Animales , Modelos Animales de Enfermedad , Distrofina/metabolismo , Humanos , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/terapia , Enfermedades Neuromusculares/complicaciones , Calidad de Vida , Respiración
3.
J Cell Physiol ; 236(9): 6407-6423, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33559206

RESUMEN

Bitter taste receptors (TAS2Rs) and their signaling elements are detected throughout the body, and bitter tastants induce a wide variety of biological responses in tissues and organs outside the mouth. However, the roles of TAS2Rs in these responses remain to be tested and established genetically. Here, we employed the CRISPR/Cas9 gene-editing technique to delete three bitter taste receptors-Tas2r143/Tas2r135/Tas2r126 (i.e., Tas2r triple knockout [TKO]) in mice. The fidelity and effectiveness of the Tas2r deletions were validated genetically at DNA and messenger RNA levels and functionally based on the tasting of TAS2R135 and TAS2R126 agonists. Bitter tastants are known to relax airways completely. However, TAS2R135 or TAS2R126 agonists either failed to induce relaxation of pre-contracted airways in wild-type mice and Tas2r TKO mice or relaxed them dose-dependently, but to the same extent in both types of mice. These results indicate that TAS2Rs are not required for bitter tastant-induced bronchodilation. The Tas2r TKO mice also provide a valuable model to resolve whether TAS2Rs mediate bitter tastant-induced responses in many other extraoral tissues.


Asunto(s)
Eliminación de Gen , Relajación Muscular , Receptores Acoplados a Proteínas G/genética , Gusto/fisiología , Animales , Secuencia de Bases , Perfilación de la Expresión Génica , Ligandos , Cloruro de Metacolina/farmacología , Ratones Noqueados , Contracción Muscular/efectos de los fármacos , Relajación Muscular/efectos de los fármacos , Músculo Liso/efectos de los fármacos , Músculo Liso/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Sistema Respiratorio/efectos de los fármacos , Sistema Respiratorio/metabolismo , Gusto/efectos de los fármacos , Lengua/efectos de los fármacos , Lengua/metabolismo
4.
Proc Natl Acad Sci U S A ; 115(11): 2788-2793, 2018 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-29453277

RESUMEN

Chronic obstructive pulmonary disease affects 10% of the worldwide population, and the leading genetic cause is α-1 antitrypsin (AAT) deficiency. Due to the complexity of the murine locus, which includes up to six Serpina1 paralogs, no genetic animal model of the disease has been successfully generated until now. Here we create a quintuple Serpina1a-e knockout using CRISPR/Cas9-mediated genome editing. The phenotype recapitulates the human disease phenotype, i.e., absence of hepatic and circulating AAT translates functionally to a reduced capacity to inhibit neutrophil elastase. With age, Serpina1 null mice develop emphysema spontaneously, which can be induced in younger mice by a lipopolysaccharide challenge. This mouse models not only AAT deficiency but also emphysema and is a relevant genetic model and not one based on developmental impairment of alveolarization or elastase administration. We anticipate that this unique model will be highly relevant not only to the preclinical development of therapeutics for AAT deficiency, but also to emphysema and smoking research.


Asunto(s)
Enfisema Pulmonar/genética , alfa 1-Antitripsina/genética , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Hígado/metabolismo , Pulmón/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Enfisema Pulmonar/metabolismo , alfa 1-Antitripsina/metabolismo
5.
Genet Med ; 22(5): 898-907, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31904026

RESUMEN

PURPOSE: Enzyme replacement therapy (ERT) with recombinant human acid-α glucosidase (rhGAA) at standard dose of 20 mg/kg every other week is insufficient to halt the long-term progression of myopathy in Pompe disease. METHODS: We conducted a retrospective study on infantile-onset Pompe disease (IPD) and late-onset Pompe disease (LOPD) patients with onset before age 5 years, ≥12 months of treatment with standard dose ERT, and rhGAA immunogenic tolerance prior to dose escalation. Long-term follow-up of up to 18 years was obtained. We obtained physical therapy, lingual strength, biochemical, and pulmonary assessments as dose was escalated. RESULTS: Eleven patients with IPD (n = 7) or LOPD (n = 4) were treated with higher doses of up to 40 mg/kg weekly. There were improvements in gross motor function measure in 9/10 patients, in lingual strength in 6/6 patients, and in pulmonary function in 4/11. Significant reductions in urinary glucose tetrasaccharide, creatine kinase, aspartate aminotransferase, and alanine aminotransferase were observed at 40 mg/kg weekly compared with lower doses (p < 0.05). No safety or immunogenicity concerns were observed at higher doses. CONCLUSION: Higher rhGAA doses are safe, improve gross motor outcomes, lingual strength, pulmonary function measures, and biochemical markers in early-onset Pompe disease, and should be considered in patients with clinical and functional decline.


Asunto(s)
Enfermedad del Almacenamiento de Glucógeno Tipo II , alfa-Glucosidasas , Niño , Preescolar , Terapia de Reemplazo Enzimático , Enfermedad del Almacenamiento de Glucógeno Tipo II/tratamiento farmacológico , Humanos , Estudios Retrospectivos , alfa-Glucosidasas/uso terapéutico
6.
Int J Mol Sci ; 21(6)2020 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-32214050

RESUMEN

Pompe disease is a glycogen storage disease caused by a deficiency in acid α-glucosidase (GAA), a hydrolase necessary for the degradation of lysosomal glycogen. This deficiency in GAA results in muscle and neuronal glycogen accumulation, which causes respiratory insufficiency. Pompe disease mouse models provide a means of assessing respiratory pathology and are important for pre-clinical studies of novel therapies that aim to treat respiratory dysfunction and improve quality of life. This review aims to compile and summarize existing manuscripts that characterize the respiratory phenotype of Pompe mouse models. Manuscripts included in this review were selected utilizing specific search terms and exclusion criteria. Analysis of these findings demonstrate that Pompe disease mouse models have respiratory physiological defects as well as pathologies in the diaphragm, tongue, higher-order respiratory control centers, phrenic and hypoglossal motor nuclei, phrenic and hypoglossal nerves, neuromuscular junctions, and airway smooth muscle. Overall, the culmination of these pathologies contributes to severe respiratory dysfunction, underscoring the importance of characterizing the respiratory phenotype while developing effective therapies for patients.


Asunto(s)
Modelos Animales de Enfermedad , Enfermedad del Almacenamiento de Glucógeno Tipo II/genética , Fenotipo , Respiración , Animales , Enfermedad del Almacenamiento de Glucógeno Tipo II/patología , Enfermedad del Almacenamiento de Glucógeno Tipo II/fisiopatología , Ratones
7.
J Inherit Metab Dis ; 42(5): 870-877, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-30993714

RESUMEN

Very-long chain acyl-CoA dehydrogenase (VLCAD) deficiency (VLCADD) is an autosomal recessive disorder of fatty acid oxidation. Fatty acids are a major source of energy during catabolic stress, so the absence of VLCAD can result in a metabolic crises and respiratory insufficiency. The etiology of this respiratory insufficiency is unclear. Thus, our aims were: (1) to characterize respiratory pathophysiology in VLCADD mice (VLCAD-/- ), and (2) to determine if AAV9-mediated gene therapy improves respiratory function. For the first aim, VLCAD-/- and wild-type (WT) mice underwent an exercise/fast "stress protocol" and awake spontaneous breathing was evaluated using whole-body plethysmography (WBP) both at baseline and during a hypercapnic respiratory challenge (FiO2 : 0.21; FiCO2 : 0.07; nitrogen balance). During hypercapnia, VLCAD -/- mice had a significantly lower frequency, tidal volume, minute ventilation, and peak inspiratory and expiratory flow, all of which indicate respiratory insufficiency. Histologically, the cardiac and respiratory muscles of stressed VLCAD -/- animals had an accumulation of intramyocellular lipids. For the second aim, a single systemic injection of AAV9-VLCAD gene therapy improved this respiratory pathology by normalizing breathing frequency and enhancing peak inspiratory flow. In addition, following gene therapy, there was a moderate reduction of lipid accumulation in the respiratory muscles. Furthermore, VLCAD protein expression was robust in cardiac and respiratory muscle. This was confirmed by immuno-staining with anti-human VLCAD antibody. In summary, stress with exercise and fasting induces respiratory insufficiency in VLCAD-/- mice and a single injection with AAV9-VLCAD gene therapy ameliorates breathing.


Asunto(s)
Acil-CoA Deshidrogenasa de Cadena Larga/deficiencia , Síndromes Congénitos de Insuficiencia de la Médula Ósea/terapia , Terapia Genética , Errores Innatos del Metabolismo Lipídico/terapia , Enfermedades Mitocondriales/terapia , Enfermedades Musculares/terapia , Insuficiencia Respiratoria/terapia , Acil-CoA Deshidrogenasa de Cadena Larga/genética , Animales , Carnitina/sangre , Síndromes Congénitos de Insuficiencia de la Médula Ósea/genética , Dependovirus/genética , Femenino , Expresión Génica , Vectores Genéticos , Metabolismo de los Lípidos , Errores Innatos del Metabolismo Lipídico/genética , Hígado/metabolismo , Masculino , Ratones , Ratones Noqueados , Enfermedades Mitocondriales/genética , Enfermedades Musculares/genética , Insuficiencia Respiratoria/etiología , Transducción Genética
8.
Int J Mol Sci ; 20(3)2019 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-30754627

RESUMEN

The authors of the recently published, "Molecular Pathways and Respiratory Involvement in Lysosomal Storage Diseases", provide an important review of the various mechanisms of lysosomal storage diseases (LSD) and how they culminate in similar clinical pathologies [...].


Asunto(s)
Enfermedad del Almacenamiento de Glucógeno Tipo II/complicaciones , Macroglosia/etiología , Macroglosia/fisiopatología , Enfermedad de la Neurona Motora/etiología , Enfermedad de la Neurona Motora/fisiopatología , Insuficiencia Respiratoria/etiología , Insuficiencia Respiratoria/fisiopatología , Humanos
9.
Am J Physiol Lung Cell Mol Physiol ; 312(6): L873-L881, 2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28336814

RESUMEN

Pompe disease is an autosomal recessive disorder caused by a deficiency of acid α-glucosidase (GAA), an enzyme responsible for hydrolyzing lysosomal glycogen. Deficiency of GAA leads to systemic glycogen accumulation in the lysosomes of skeletal muscle, motor neurons, and smooth muscle. Skeletal muscle and motor neuron pathology are known to contribute to respiratory insufficiency in Pompe disease, but the role of airway pathology has not been evaluated. Here we propose that GAA enzyme deficiency disrupts the function of the trachea and bronchi and this lower airway pathology contributes to respiratory insufficiency in Pompe disease. Using an established mouse model of Pompe disease, the Gaa-/- mouse, we compared histology, pulmonary mechanics, airway smooth muscle (ASM) function, and calcium signaling between Gaa-/- and age-matched wild-type (WT) mice. Lysosomal glycogen accumulation was observed in the smooth muscle of both the bronchi and the trachea in Gaa-/- but not WT mice. Furthermore, Gaa-/- mice had hyporesponsive airway resistance and bronchial ring contraction to the bronchoconstrictive agents methacholine (MCh) and potassium chloride (KCl) and to a bronchodilator (albuterol). Finally, calcium signaling during bronchiolar smooth muscle contraction was impaired in Gaa-/- mice indicating impaired extracellular calcium influx. We conclude that GAA enzyme deficiency leads to glycogen accumulation in the trachea and bronchi and impairs the ability of lower ASM to regulate calcium and respond appropriately to bronchodilator or constrictors. Accordingly, ASM dysfunction may contribute to respiratory impairments in Pompe disease.


Asunto(s)
Enfermedad del Almacenamiento de Glucógeno Tipo II/enzimología , Enfermedad del Almacenamiento de Glucógeno Tipo II/fisiopatología , Pulmón/enzimología , Pulmón/patología , Músculo Esquelético/enzimología , Músculo Esquelético/fisiopatología , alfa-Glucosidasas/metabolismo , Albuterol/farmacología , Animales , Bronquios/efectos de los fármacos , Bronquios/fisiopatología , Señalización del Calcio/efectos de los fármacos , Espacio Extracelular/metabolismo , Glucógeno/metabolismo , Enfermedad del Almacenamiento de Glucógeno Tipo II/patología , Pulmón/efectos de los fármacos , Pulmón/fisiopatología , Cloruro de Metacolina/farmacología , Ratones , Contracción Muscular/efectos de los fármacos , Músculo Esquelético/efectos de los fármacos , Cloruro de Potasio/farmacología , Tráquea/efectos de los fármacos , Tráquea/fisiopatología
10.
Hum Mol Genet ; 24(3): 625-36, 2015 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-25217571

RESUMEN

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.


Asunto(s)
Enfermedad del Almacenamiento de Glucógeno Tipo II/patología , Glicoproteínas de Membrana/metabolismo , Músculo Esquelético/patología , Unión Neuromuscular/patología , Nervio Frénico/patología , Animales , Diafragma/metabolismo , Diafragma/patología , Modelos Animales de Enfermedad , Enfermedad del Almacenamiento de Glucógeno Tipo II/genética , Enfermedad del Almacenamiento de Glucógeno Tipo II/metabolismo , Humanos , Glicoproteínas de Membrana/genética , Ratones , Ratones Noqueados , Músculo Esquelético/metabolismo , Unión Neuromuscular/metabolismo , Nervio Frénico/metabolismo , Tibia/metabolismo , Tibia/patología
11.
Ann Neurol ; 79(4): 687-700, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26891182

RESUMEN

OBJECTIVE: Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease characterized by loss of motor neurons, resulting in progressive muscle weakness, paralysis, and death within 5 years of diagnosis. About 10% of cases are inherited, of which 20% are due to mutations in the superoxide dismutase 1 (SOD1) gene. Riluzole, the only US Food and Drug Administration-approved ALS drug, prolongs survival by only a few months. Experiments in transgenic ALS mouse models have shown decreasing levels of mutant SOD1 protein as a potential therapeutic approach. We sought to develop an efficient adeno-associated virus (AAV)-mediated RNAi gene therapy for ALS. METHODS: A single-stranded AAV9 vector encoding an artificial microRNA against human SOD1 was injected into the cerebral lateral ventricles of neonatal SOD1(G93A) mice, and impact on disease progression and survival was assessed. RESULTS: This therapy extended median survival by 50% and delayed hindlimb paralysis, with animals remaining ambulatory until the humane endpoint, which was due to rapid body weight loss. AAV9-treated SOD1(G93A) mice showed reduction of mutant human SOD1 mRNA levels in upper and lower motor neurons and significant improvements in multiple parameters including the numbers of spinal motor neurons, diameter of ventral root axons, and extent of neuroinflammation in the SOD1(G93A) spinal cord. Mice also showed previously unexplored changes in pulmonary function, with AAV9-treated SOD1(G93A) mice displaying a phenotype reminiscent of patient pathophysiology. INTERPRETATION: These studies clearly demonstrate that an AAV9-delivered SOD1-specific artificial microRNA is an effective and translatable therapeutic approach for ALS.


Asunto(s)
Esclerosis Amiotrófica Lateral/terapia , Dependovirus , Terapia Genética/métodos , Vectores Genéticos , MicroARNs/uso terapéutico , Superóxido Dismutasa/uso terapéutico , Animales , Animales Recién Nacidos , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Inyecciones Intraventriculares , Ventrículos Laterales , Ratones , Ratones Transgénicos , Superóxido Dismutasa-1
12.
J Neurophysiol ; 115(3): 1372-80, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26683067

RESUMEN

Power spectral analyses of electrical signals from respiratory nerves reveal prominent oscillations above the primary rate of breathing. Acute exposure to intermittent hypoxia can induce a form of neuroplasticity known as long-term facilitation (LTF), in which inspiratory burst amplitude is persistently elevated. Most evidence indicates that the mechanisms of LTF are postsynaptic and also that high-frequency oscillations within the power spectrum show coherence across different respiratory nerves. Since the most logical interpretation of this coherence is that a shared presynaptic mechanism is responsible, we hypothesized that high-frequency spectral content would be unchanged during LTF. Recordings of inspiratory hypoglossal (XII) activity were made from anesthetized, vagotomized, and ventilated 129/SVE mice. When arterial O2 saturation (SaO2) was maintained >96%, the XII power spectrum and burst amplitude were unchanged for 90 min. Three, 1-min hypoxic episodes (SaO2 = 50 ± 10%), however, caused a persistent (>60 min) and robust (>400% baseline) increase in burst amplitude. Spectral analyses revealed a rightward shift of the signal content during LTF, with sustained increases in content above ∼125 Hz following intermittent hypoxia and reductions in power at lower frequencies. Changes in the spectral content during LTF were qualitatively similar to what occurred during the acute hypoxic response. We conclude that high-frequency content increases during XII LTF in this experimental preparation; this may indicate that intermittent hypoxia-induced plasticity in the premotor network contributes to expression of XII LTF.


Asunto(s)
Nervio Hipogloso/fisiología , Hipoxia/fisiopatología , Potenciación a Largo Plazo , Animales , Nervio Hipogloso/fisiopatología , Masculino , Ratones , Potenciales Sinápticos
13.
Am J Respir Cell Mol Biol ; 53(3): 326-35, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25569118

RESUMEN

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.


Asunto(s)
Enfermedad del Almacenamiento de Glucógeno Tipo II/tratamiento farmacológico , Isoxazoles/farmacología , Respiración/efectos de los fármacos , Fármacos del Sistema Respiratorio/farmacología , Animales , Tronco Encefálico/patología , Evaluación Preclínica de Medicamentos , Enfermedad del Almacenamiento de Glucógeno Tipo II/fisiopatología , Isoxazoles/uso terapéutico , Ratones de la Cepa 129 , Ratones Noqueados , Actividad Motora/efectos de los fármacos , Nervio Frénico/efectos de los fármacos , Nervio Frénico/fisiopatología , Fármacos del Sistema Respiratorio/uso terapéutico
14.
Mol Ther ; 22(4): 702-12, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24336173

RESUMEN

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.


Asunto(s)
Terapia Genética , Enfermedad del Almacenamiento de Glucógeno Tipo II/genética , Enfermedad del Almacenamiento de Glucógeno Tipo II/terapia , Neuronas Motoras/metabolismo , alfa-Glucosidasas/genética , Animales , Dependovirus/genética , Regulación Enzimológica de la Expresión Génica , Técnicas de Transferencia de Gen , Glucógeno , Enfermedad del Almacenamiento de Glucógeno Tipo II/patología , Humanos , Inyecciones Intramusculares , Ratones , Neuronas Motoras/patología , Músculo Esquelético/metabolismo , Miofibrillas/genética , Miofibrillas/metabolismo , Regiones Promotoras Genéticas , alfa-Glucosidasas/biosíntesis
15.
J Community Health ; 40(3): 409-13, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25245161

RESUMEN

Through a QI project at a tertiary referral pediatric pulmonary center, our objective was to establish a methodical approach to identify and engage smoking parents of children with chronic lung disease in a smoking cessation program. We hypothesized that smoking caregivers of children with chronic lung disease would be more motivated to enroll in a smoking cessation program when referred from tertiary pediatric pulmonary center. We assessed smoking habits and interest in quitting of parents with surveys. Parents ready to quit within 30 days were referred to the Florida Quitline from clinic. Pulmonary function tests, exacerbations, hospitalizations and need for prednisone or antibiotics were obtained from the patient charts and surveys. Follow-up two to 6 months later assessed the quit rate and child's clinical well-being and lung function. A standard mechanism to identify caregivers who smoked was established by engaging our medical assistants through a prompt in our EMR system. Out of those caregivers who were identified as smokers and accompanied their children to clinic, 52% were interested in a referral to the Florida Quitline. Out of those, only 12% successfully completed the program and ceased to smoke. The Florida Quitline was unable to reach the majority of parents who were referred to them. The majority of those referred to the Ouitline were not successfully contacted or enrolled in the program. The current procedure for referring and enrolling individuals to the Quitline is not effective for our population, but compares to the national average.


Asunto(s)
Enfermedades Pulmonares/epidemiología , Padres , Derivación y Consulta/organización & administración , Cese del Hábito de Fumar/métodos , Fumar/epidemiología , Contaminación por Humo de Tabaco/prevención & control , Adolescente , Corticoesteroides/administración & dosificación , Adulto , Antibacterianos/administración & dosificación , Asma/epidemiología , Cuidadores , Niño , Preescolar , Enfermedad Crónica , Fibrosis Quística/epidemiología , Femenino , Florida/epidemiología , Líneas Directas , Humanos , Lactante , Intención , Masculino , Persona de Mediana Edad , Pruebas de Función Respiratoria , Adulto Joven
16.
Mol Ther ; 21(9): 1661-7, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23732990

RESUMEN

Pompe disease is a neuromuscular disease resulting from deficiency in acid α-glucosidase (GAA), results in cardiac, skeletal muscle, and central nervous system (CNS) pathology. Enzyme replacement therapy (ERT) has been shown to partially correct cardiac and skeletal muscle dysfunction. However, ERT does not cross the blood-brain barrier and progressive CNS pathology ensues. We tested the hypothesis that intrapleural administration of recombinant adeno-associated virus (rAAV9)-GAA driven by a cytomegalovirus (CMV) or desmin (DES) promoter would improve cardiac and respiratory function in Gaa(-/-) mice through a direct effect and retrograde transport to motoneurons. Cardiac magnetic resonance imaging revealed significant improvement in ejection fraction in rAAV9-GAA-treated animals. Inspiratory phrenic and diaphragm activity was examined at baseline and during hypercapnic respiratory challenge. Mice treated with AAV9 had greater relative inspiratory burst amplitude during baseline conditions when compared with Gaa(-/-). In addition, efferent phrenic burst amplitude was significantly correlated with diaphragm activity in both AAV9-DES and AAV9-CMV groups but not in Gaa(-/-). This is the first study to indicate improvements in cardiac, skeletal muscle, and respiratory neural output following rAAV administration in Pompe disease. These results further implicate a role for the CNS in Pompe disease pathology and the critical need to target the neurologic aspects in developing therapeutic strategies.


Asunto(s)
Dependovirus/genética , Enfermedad del Almacenamiento de Glucógeno Tipo II/fisiopatología , Enfermedad del Almacenamiento de Glucógeno Tipo II/terapia , Corazón/fisiología , Nervio Frénico/fisiología , Músculos Respiratorios/fisiología , alfa-Glucosidasas/genética , Animales , Dependovirus/metabolismo , Diafragma/fisiología , Modelos Animales de Enfermedad , Vectores Genéticos , Enfermedad del Almacenamiento de Glucógeno Tipo II/genética , Humanos , Ratones , Músculo Esquelético/patología , Músculo Esquelético/fisiología , Miocardio/metabolismo , Miocardio/patología , Pleura , Distribución Aleatoria , Médula Espinal/metabolismo , Transducción Genética , alfa-Glucosidasas/metabolismo
17.
Respir Physiol Neurobiol ; 326: 104282, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38782084

RESUMEN

Duchenne muscular dystrophy (DMD) is the most common X-linked disease. DMD is caused by a lack of dystrophin, a critical structural protein in striated muscle. Dystrophin deficiency leads to inflammation, fibrosis, and muscle atrophy. Boys with DMD have progressive muscle weakness within the diaphragm that results in respiratory failure in the 2nd or 3rd decade of life. The most common DMD mouse model - the mdx mouse - is not sufficient for evaluating genetic medicines that specifically target the human DMD (hDMD) gene sequence. Therefore, a novel transgenic mouse carrying the hDMD gene with an exon 52 deletion was created (hDMDΔ52;mdx). We characterized the respiratory function and pathology in this model using whole body plethysmography, histology, and immunohistochemistry. At 6-months-old, hDMDΔ52;mdx mice have reduced maximal respiration, neuromuscular junction pathology, and fibrosis throughout the diaphragm, which worsens at 12-months-old. In conclusion, the hDMDΔ52;mdx exhibits moderate respiratory pathology, and serves as a relevant animal model to study the impact of novel genetic therapies, including gene editing, on respiratory function.


Asunto(s)
Modelos Animales de Enfermedad , Ratones Transgénicos , Distrofia Muscular de Duchenne , Animales , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/patología , Distrofia Muscular de Duchenne/fisiopatología , Ratones , Humanos , Masculino , Distrofina/genética , Distrofina/deficiencia , Ratones Endogámicos mdx , Diafragma/fisiopatología , Diafragma/patología , Insuficiencia Respiratoria/etiología , Unión Neuromuscular/patología , Unión Neuromuscular/metabolismo , Ratones Endogámicos C57BL
18.
HGG Adv ; 5(3): 100288, 2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38566418

RESUMEN

Biallelic loss-of-function variants in the MUSK gene result in two allelic disorders: (1) congenital myasthenic syndrome (CMS; OMIM: 616325), a neuromuscular disorder that has a range of severity from severe neonatal-onset weakness to mild adult-onset weakness, and (2) fetal akinesia deformation sequence (OMIM: 208150), a form of pregnancy loss characterized by severe muscle weakness in the fetus. The MUSK gene codes for muscle-specific kinase (MuSK), a receptor tyrosine kinase involved in the development of the neuromuscular junction. Here, we report a case of neonatal-onset MUSK-related CMS in a patient harboring compound heterozygous deletions in the MUSK gene, including (1) a deletion of exons 2-3 leading to an in-frame MuSK protein lacking the immunoglobulin 1 (Ig1) domain and (2) a deletion of exons 7-11 leading to an out-of-frame, truncated MuSK protein. Individual domains of the MuSK protein have been elucidated structurally; however, a complete MuSK structure generated by machine learning algorithms has clear inaccuracies. We modify a predicted AlphaFold structure and integrate previously reported domain-specific structural data to suggest a MuSK protein that dimerizes in two locations (Ig1 and the transmembrane domain). We analyze known pathogenic variants in MUSK to discover domain-specific genotype-phenotype correlations; variants that lead to a loss of protein expression, disruption of the Ig1 domain, or Dok-7 binding are associated with the most severe phenotypes. A conceptual model is provided to explain the severe phenotypes seen in Ig1 variants and the poor response of our patient to pyridostigmine.

19.
Hum Mol Genet ; 20(R1): R61-8, 2011 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-21518733

RESUMEN

Pompe disease is an autosomal recessive metabolic myopathy caused by the deficiency of the lysosomal enzyme acid alpha-glucosidase and results in cellular lysosomal and cytoplasmic glycogen accumulation. A wide spectrum of disease exists from hypotonia and severe cardiac hypertrophy in the first few months of life due to severe mutations to a milder form with the onset of symptoms in adulthood. In either condition, the involvement of several systems leads to progressive weakness and disability. In early-onset severe cases, the natural history is characteristically cardiorespiratory failure and death in the first year of life. Since the advent of enzyme replacement therapy (ERT), the clinical outcomes have improved. However, it has become apparent that a new natural history is being defined in which some patients have substantial improvement following ERT, while others develop chronic disability reminiscent of the late-onset disease. In order to improve on the current clinical outcomes in Pompe patients with diminished clinical response to ERT, we sought to address the cause and potential for the treatment of disease manifestations which are not amenable to ERT. In this review, we will focus on the preclinical studies that are relevant to the development of a gene therapy strategy for Pompe disease, and have led to the first clinical trial of recombinant adeno-associated virus-mediated gene-based therapy for Pompe disease. We will cover the preliminary laboratory studies and rationale for a clinical trial, which is based on the treatment of the high rate of respiratory failure in the early-onset patients receiving ERT.


Asunto(s)
Dependovirus/genética , Terapia Genética/métodos , Enfermedad del Almacenamiento de Glucógeno Tipo II/terapia , Ensayos Clínicos como Asunto , Terapia de Reemplazo Enzimático , Vectores Genéticos/administración & dosificación , Glucógeno/metabolismo , Enfermedad del Almacenamiento de Glucógeno Tipo II/inmunología , Enfermedad del Almacenamiento de Glucógeno Tipo II/patología , Humanos , Resultado del Tratamiento
20.
Respir Med Case Rep ; 43: 101842, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37077236

RESUMEN

Congenital lobar emphysema (CLE) is caused by airway defects resulting in air trapping and hyperinflation of the affected lobe. Case reports of families affected with CLE imply a genetic etiology. However, the genetic contributions have not been well-described. We present a case of CLE in a monozygotic twin brother with respiratory distress diagnosed with right upper lobe (RUL) CLE and treated with a lobectomy. His asymptomatic twin brother was screened prophylactically, found to have RUL CLE and subsequently underwent a lobectomy. Our report provides further evidence supporting the genetic predisposition and potential benefit of early screening for CLE in comparable scenarios.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA