Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
FASEB J ; 35(6): e21628, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33982338

RESUMEN

Duchenne muscular dystrophy (DMD) is a lethal genetic muscle disorder caused by recessive mutations in dystrophin gene, affecting 1/3000 males. Gene therapy has been proven to ameliorate dystrophic pathology. To investigate therapeutic benefits from long-term effect of human mini-dystrophin and functional outcomes, transgenic mdx mice (Tg-mdx) containing a single copy of human mini-dystrophin (∆hDys3849) gene, five rods (Rods1-2, Rods22-24), and two hinges (H1 and H4) driven by a truncated creatine-kinase promoter (dMCK) in a recombinant adeno-associated viral vector (rAAV) backbone, were generated and used to determine gene expression and improvement of muscle function. Human mini-dystrophin gene expression was found in a majority of the skeletal muscles, but no expression in cardiac muscle. Dystrophin-associated glycoproteins (DAGs) such as sarcoglycans and nNOS were restored at the sarcolemma and coincided with human mini-dystrophin gene expression at the ages of 6, 10, and 20 months; Morphology of dystrophic muscle expressing the human mini-dystrophin gene was improved and central nuclei were reduced. Myofiber membrane integrity was improved by Evans blue dye test. Improvement in treadmill running and grip force was observed in transgenic mice at 6 months. Tetanic force and specific force of tibialis anterior (TA) muscle were significantly increased at the ages of 6, 10, and 20 months. Pseudohypertrophy was not found in TA muscle at 10 and 20 months when compared with wild-type C57 (WT) group. This study demonstrated that the long-term effects of human mini-dystrophin effectively ameliorated pathology and improved the functions of the dystrophic muscles in the transgenic DMD mouse model.


Asunto(s)
Distrofina/metabolismo , Terapia Genética , Músculo Esquelético/fisiología , Distrofia Muscular Animal/terapia , Distrofia Muscular de Duchenne/terapia , Animales , Distrofina/genética , Humanos , Ratones , Ratones Endogámicos mdx , Ratones Transgénicos , Músculo Esquelético/citología , Distrofia Muscular Animal/etiología , Distrofia Muscular Animal/patología , Distrofia Muscular de Duchenne/etiología , Distrofia Muscular de Duchenne/patología
2.
Gene Ther ; 27(5): 237-244, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31819202

RESUMEN

Recombinant adeno-associated viral (AAV) vectors are frequently used to deliver DNA into cells and are currently the leading platform for therapeutic gene delivery in humans. Presently, there is a need for optimized AAV vectors with improved transduction efficiencies in target tissues. In these studies, an engineered albumin-binding consensus domain (ABDCon) peptide was incorporated into the AAV9 capsid via fusion to the N-terminus of the AAV9 VP2 capsid protein to generate a variant AAV9 capsid with albumin-binding properties. The variant capsid, called AAV9-ABDCon, formed viable genome-containing vector particles and exhibited binding to human serum albumin. The AAV9 capsid, on the other hand, was not found to bind to human serum albumin by the methods used in this study. In C57BL/6J mice, AAV9-ABDCon achieved significantly higher levels of liver transduction compared with AAV9 following intravenous administration. These findings show that incorporation of the ABDCon peptide into the capsid VP2 N-terminus may be a potential method to augment AAV-mediated liver-directed gene delivery.


Asunto(s)
Cápside , Dependovirus , Administración Intravenosa , Animales , Proteínas de la Cápside/genética , Dependovirus/genética , Vectores Genéticos/genética , Humanos , Hígado , Ratones , Ratones Endogámicos C57BL , Albúmina Sérica/genética , Transducción Genética
3.
Mol Ther ; 26(4): 1109-1117, 2018 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-29503194

RESUMEN

Growth and differentiation factor 11 (GDF11; BMP11) is a circulating cytokine in the transforming growth factor beta (TGF-ß) superfamily. Treatment with recombinant GDF11 (rGDF11) protein has previously been shown to reverse skeletal muscle dysfunction in aged mice. However, the actions of GDF11 in skeletal muscle are still not fully understood. Because GDF11 activates the TGF-ß-SMAD2/3 pathway, we hypothesized that GDF11 overexpression would inhibit skeletal muscle growth. To test this hypothesis, we generated recombinant adeno-associated virus serotype 9 (AAV9) vectors harboring the gene for either human GDF11 (AAV9-GDF11) or human IgG1 Fc-fused GDF11 propeptide (AAV9-GDF11Pro-Fc-1) to study the effects of GDF11 overexpression or blockade on skeletal muscle growth and function in vivo. After intravenous administration of AAV9-GDF11 into neonatal C57BL/6J mice, we observed sustained limb muscle growth inhibition along with reductions in forelimb grip strength and treadmill running endurance at 16 weeks. Conversely, treatment with AAV9-GDF11Pro-Fc-1 led to increased limb muscle mass and forelimb grip strength after 28 weeks, although a difference in the total body mass/muscle mass ratio was not observed between treatment and control groups. In sum, our results suggest GDF11 overexpression has an inhibitory effect on skeletal muscle growth.


Asunto(s)
Proteínas Morfogenéticas Óseas/genética , Dependovirus/genética , Expresión Génica , Vectores Genéticos/genética , Factores de Diferenciación de Crecimiento/genética , Músculo Esquelético/metabolismo , Transgenes , Animales , Orden Génico , Técnicas de Transferencia de Gen , Humanos , Inmunohistoquímica , Ratones , Miocardio/metabolismo , Miocardio/patología
4.
Am J Pathol ; 187(2): 431-440, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28107841

RESUMEN

Agrin is a basement membrane-specific proteoglycan that can regulate orientation of cytoskeleton proteins and improve function of dystrophic skeletal muscle. In skeletal muscle, agrin binds with high affinity to laminin(s) and α-dystroglycan (α-DG), an integral part of the dystrophin-glycoprotein complex. Miniaturized forms of agrin (mAgrin) have been shown to ameliorate disease pathology in a laminin-α2 knockout mouse model of muscular dystrophy, acting as a link between α-DG and laminin(s). Here, we test whether mAgrin might also improve pathologies associated with FKRP-related dystroglycanopathies, another form of muscular dystrophy characterized by weak interactions between muscle and basement membranes. We demonstrate in vitro that mAgrin enhances laminin binding to primary myoblasts and fibroblasts from an FKRP mutant mouse model and that this enhancement is abrogated when mAgrin is in molar excess relative to laminin. However, in vivo delivery of mAgrin via adeno-associated virus (AAV) into FKRP mutant mice was unable to improve dystrophic phenotypes, both histologically and functionally. These results likely reflect insufficient binding of mAgrin to hypoglycosylated α-DG on muscle fibers and possibly abrogation of binding from molar excess of overexpressed AAV-delivered mAgrin. Further exploration of mAgrin modification is necessary to strengthen its binding to other membrane components, including hypoglycosylated α-DG, for potential therapeutic applications.


Asunto(s)
Agrina/genética , Terapia Genética/métodos , Distrofia Muscular Animal/terapia , Agrina/metabolismo , Animales , Western Blotting , Dependovirus , Inmunohistoquímica , Laminina/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Distrofia Muscular de Cinturas , Distrofia Muscular Animal/patología , Fenotipo , Unión Proteica
5.
Mol Ther ; 23(5): 866-874, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25676679

RESUMEN

Diabetes poses a substantial burden to society as it can lead to serious complications and premature death. The number of cases continues to increase worldwide. Two major causes of diabetes are insulin resistance and insulin insufficiency. Currently, there are few antidiabetic drugs available that can preserve or protect ß-cell function to overcome insulin insufficiency in diabetes. We describe a therapeutic strategy to preserve ß-cell function by overexpression of follistatin (FST) using an AAV vector (AAV8-Ins-FST) in diabetic mouse model. Overexpression of FST in the pancreas of db/db mouse increased ß-cell islet mass, decreased fasting glucose level, alleviated diabetic symptoms, and essentially doubled lifespan of the treated mice. The observed islet enlargement was attributed to ß-cell proliferation as a result of bioneutralization of myostatin and activin by FST. Overall, our study indicates overexpression of FST in the diabetic pancreas preserves ß-cell function by promoting ß-cell proliferation, opening up a new therapeutic avenue for the treatment of diabetes.


Asunto(s)
Folistatina/genética , Expresión Génica , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Animales , Biomarcadores , Proliferación Celular , Dependovirus/clasificación , Dependovirus/genética , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/terapia , Modelos Animales de Enfermedad , Folistatina/metabolismo , Terapia Genética , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Humanos , Inmunohistoquímica , Insulina/sangre , Islotes Pancreáticos/anatomía & histología , Islotes Pancreáticos/metabolismo , Ligandos , Masculino , Ratones , Fenotipo , Fosfatidilinositol 3-Quinasas/metabolismo , Unión Proteica , Proteínas Proto-Oncogénicas c-akt/metabolismo , Serogrupo , Transducción de Señal , Proteínas Smad/metabolismo , Transducción Genética , Transgenes
6.
Am J Physiol Cell Physiol ; 309(11): C724-35, 2015 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-26333599

RESUMEN

The commercial availability of multiphoton microscope systems has nurtured the growth of intravital microscopy as a powerful technique for evaluating cell biology in the relevant context of living animals. In parallel, new fluorescent protein (FP) biosensors have become available that enable studies of the function of a wide range of proteins in living cells. Biosensor probes that exploit Förster resonance energy transfer (FRET) are among the most sensitive indicators of an array of cellular processes. However, differences between one-photon and two-photon excitation (2PE) microscopy are such that measuring FRET by 2PE in the intravital setting remains challenging. Here, we describe an approach that simplifies the use of FRET-based biosensors in intravital 2PE microscopy. Based on a systematic comparison of many different FPs, we identified the monomeric (m) FPs mTurquoise and mVenus as particularly well suited for intravital 2PE FRET studies, enabling the ratiometric measurements from linked FRET probes using a pair of experimental images collected simultaneously. The behavior of the FPs is validated by fluorescence lifetime and sensitized emission measurements of a set of FRET standards. The approach is demonstrated using a modified version of the AKAR protein kinase A biosensor, first in cells in culture, and then in hepatocytes in the liver of living mice. The approach is compatible with the most common 2PE microscope configurations and should be applicable to a variety of different FRET probes.


Asunto(s)
Técnicas Biosensibles/métodos , Transferencia Resonante de Energía de Fluorescencia/métodos , Microscopía de Fluorescencia por Excitación Multifotónica/métodos , Miocitos Cardíacos/química , Animales , Células Cultivadas , Colorantes Fluorescentes/análisis , Células HEK293 , Humanos , Proteínas Luminiscentes/análisis , Ratones , Microscopía Confocal/métodos
7.
Mol Ther ; 22(11): 1890-9, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25048216

RESUMEN

Mutations in fukutin-related protein (FKRP) gene cause a wide spectrum of disease phenotypes including the mild limb-girdle muscular dystrophy 2I (LGMD2I), the severe Walker-Warburg syndrome, and muscle-eye-brain disease. FKRP deficiency results in α-dystroglycan (α-DG) hypoglycosylation in the muscle and heart, which is a biochemical hallmark of dystroglycanopathies. To study gene replacement therapy, we generated and characterized a new mouse model of LGMD2I harboring the human mutation leucine 276 to isoleucine (L276I) in the mouse alleles. The homozygous knock-in mice (L276I(KI)) mimic the classic late onset phenotype of LGMD2I in both skeletal and cardiac muscles. Systemic delivery of human FKRP gene by AAV9 vector in the L276I(KI) mice, at either neonatal age or at the age of 9 months, rendered body wide FKRP expression and restored glycosylation of α-DG in both skeletal and cardiac muscles. FKRP gene therapy ameliorated dystrophic pathology and cardiomyopathy such as muscle degeneration, fibrosis, and myofiber membrane leakage, resulting in restoration of muscle and heart contractile functions. Thus, these results demonstrated that the treatment based on FKRP gene replacement was effective.


Asunto(s)
Terapia Genética/métodos , Corazón/fisiopatología , Distrofia Muscular de Cinturas/terapia , Proteínas/genética , Animales , Modelos Animales de Enfermedad , Técnicas de Inactivación de Genes , Humanos , Ratones , Distrofia Muscular de Cinturas/genética , Distrofia Muscular de Cinturas/fisiopatología , Distrofia Muscular Animal/terapia , Pentosiltransferasa
8.
Mol Ther ; 21(10): 1832-40, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23817215

RESUMEN

Mutations in the FKRP gene are associated with a wide range of muscular dystrophies from mild limb-girdle muscular dystrophy (LGMD) 2I to severe Walker-Warburg syndrome and muscle-eye-brain disease. The characteristic biochemical feature of these diseases is the hypoglycosylation of α-dystroglycan (α-DG). Currently there is no effective treatment available. In this study, we examined the adeno-associated virus serotype 9 vector (AAV9)-mediated gene therapy in the FKRP mutant mouse model with a proline to leucine missense mutation (P448L). Our results showed that intraperitoneal administration of AAV9-FKRP resulted in systemic FKRP expression in all striated muscles examined with the highest levels in cardiac muscle. Consistent with our previous observations, FKRP protein is localized in the Golgi apparatus in myofibers. Expression of FKRP consequently restored functional glycosylation of α-DG in the skeletal and cardiac muscles. Significant improvement in dystrophic pathology, serum creatine kinase levels and muscle function was observed. Only limited FKRP transgene expression was detected in kidney and liver with no detectable toxicity. Our results provided evidence for the utility of AAV-mediated gene replacement therapy for FKRP-related muscular dystrophies.


Asunto(s)
Dependovirus/genética , Distroglicanos/metabolismo , Terapia Genética/métodos , Músculo Esquelético/fisiología , Distrofia Muscular de Cinturas/terapia , Distrofia Muscular Animal/terapia , Proteínas/genética , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Vectores Genéticos , Glicosilación , Inyecciones Intraperitoneales , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Distrofia Muscular de Cinturas/metabolismo , Distrofia Muscular de Cinturas/fisiopatología , Pentosiltransferasa , Proteínas/metabolismo , Transferasas , Transgenes
9.
Mol Ther ; 20(4): 727-35, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22314291

RESUMEN

Muscular dystrophies (MDs) are caused by genetic mutations in over 30 different genes, many of which encode for proteins essential for the integrity of muscle cell structure and membrane. Their deficiencies cause the muscle vulnerable to mechanical and biochemical damages, leading to membrane leakage, dystrophic pathology, and eventual loss of muscle cells. Recent studies report that MG53, a muscle-specific TRIM-family protein, plays an essential role in sarcolemmal membrane repair. Here, we show that systemic delivery and muscle-specific overexpression of human MG53 gene by recombinant adeno-associated virus (AAV) vectors enhanced membrane repair, ameliorated pathology, and improved muscle and heart functions in δ-sarcoglycan (δ-SG)-deficient TO-2 hamsters, an animal model of MD and congestive heart failure. In addition, MG53 overexpression increased dysferlin level and facilitated its trafficking to muscle membrane through participation of caveolin-3. MG53 also protected muscle cells by activating cell survival kinases, such as Akt, extracellular signal-regulated kinases (ERK1/2), and glycogen synthase kinase-3ß (GSK-3ß) and inhibiting proapoptotic protein Bax. Our results suggest that enhancing the muscle membrane repair machinery could be a novel therapeutic approach for MD and cardiomyopathy, as demonstrated here in the limb girdle MD (LGMD) 2F model.


Asunto(s)
Proteínas Portadoras/metabolismo , Terapia Genética/métodos , Insuficiencia Cardíaca/terapia , Distrofias Musculares/terapia , Sarcoglicanos/deficiencia , Animales , Proteínas Portadoras/genética , Caveolina 3/genética , Caveolina 3/metabolismo , Cricetinae , Dependovirus/genética , Insuficiencia Cardíaca/metabolismo , Humanos , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Distrofias Musculares/metabolismo , Proteínas de Motivos Tripartitos
10.
Hepat Med ; 15: 165-183, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37822344

RESUMEN

Purpose: In this study, our primary aim is to analyze the genetic expression feature and analyze specific Traditional Chinese medicine (TCM) constitution distribution in non-alcoholic fatty liver disease (NAFLD) and reveal the metabolic characteristic of NAFLD. Materials and Methods: For revealing genetic features, we obtained the gene expression data from the Gene Expression Omnibus (GEO) database of the National Center for Biotechnology Information (NCBI). The genetic data on NAFLD were analyzed by identifying differentially expressed genes (DEGs), associated pathways, co-expressed genetic networks, and gene set enrichment function. Concurrently, we assessed specific constitution distributions among local NAFLD patients through established TCM constitution models and determined the independent variable, including specific constitution to the NAFLD via the regression analyses. Results: The analyses on GEO datasets showed that simple steatosis in NAFLD is strongly associated with HOMA-insulin resistance (HOMA-IR). Analyses of GEO datasets revealed significantly altered genetic expression profiles between NAFLD and normal populations. For TCM constitution analyses, we demonstrated a decline in yin-yang harmony (YYH) and yang-asthenia (YAAC) constitution, whereas there was an increase in qi-stagnation (QSC) and phlegm-dampness (PDC) in NAFLD. The binary logistic regression analysis indicated that besides other metabolic parameters, YYH, qi asthenia (QAC), YYAC, and yin-asthenia (YAC) were the independent variables of NAFLD, while YAC was the independent variables of T2D. The multilinear regression analyses suggested that NAFLD, DM, BMI, waist, TC, TG, hypertension, ALT, AST, and YAC were the significant determinators of the FPG. Conclusion: This study presents a relatively comprehensive metabolic profile in steatosis of NAFLD, revealed by significant genetic expression feature alterations and different TCM constitution distribution in NAFLD. Through this method, the study intends to associate the genetic feature with the phenotype of TCM constitution. The results could be applied to assist integrative medicine research in exploring the appropriate personalized approaches for NAFLD.

11.
Am J Pathol ; 178(1): 261-72, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21224063

RESUMEN

Limb-girdle muscular dystrophy 2I (LGMD2I) is caused by mutations in the fukutin-related protein (FKRP) gene. Unlike its severe allelic forms, LGMD2I usually involves slower onset and milder course without defects in the central nervous system. The lack of viable animal models that closely recapitulate LGMD2I clinical phenotypes led us to use RNA interference technology to knock down FKRP expression via postnatal gene delivery so as to circumvent embryonic lethality. Specifically, an adeno-associated viral vector was used to deliver short hairpin (shRNA) genes to healthy ICR mice. Adeno-associated viral vectors expressing a single shRNA or two different shRNAs were injected one time into the hind limb muscles. We showed that FKRP expression at 10 months postinjection was reduced by about 50% with a single shRNA and by 75% with the dual shRNA cassette. Dual-cassette injection also reduced a-dystroglycan glycosylation and its affinity to laminin by up to 70% and induced α-dystrophic pathology, including fibrosis and central nucleation, in more than 50% of the myofibers at 10 months after injection. These results suggest that the reduction of approximately or more than 75% of the normal level of FKRP expression induces chronic dystrophic phenotypes in skeletal muscles. Furthermore, the restoration of about 25% of the normal FKRP level could be sufficient for LGMD2I therapy to correct the genetic deficiency effectively and prevent dystrophic pathology.


Asunto(s)
Técnicas de Silenciamiento del Gen/métodos , Músculo Esquelético/patología , Distrofia Muscular de Cinturas/genética , Proteínas/genética , Interferencia de ARN , Adenoviridae , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Distroglicanos/metabolismo , Vectores Genéticos , Glicosilación , Ratones , Ratones Endogámicos ICR , Músculo Esquelético/metabolismo , Distrofia Muscular de Cinturas/metabolismo , Distrofia Muscular de Cinturas/patología , Pentosiltransferasa , ARN Interferente Pequeño/genética , Transferasas
12.
Proc Natl Acad Sci U S A ; 106(10): 3946-51, 2009 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-19234115

RESUMEN

To engineer gene vectors that target striated muscles after systemic delivery, we constructed a random library of adeno-associated virus (AAV) by shuffling the capsid genes of AAV serotypes 1 to 9, and screened for muscle-targeting capsids by direct in vivo panning after tail vein injection in mice. After 2 rounds of in vivo selection, a capsid gene named M41 was retrieved mainly based on its high frequency in the muscle and low frequency in the liver. Structural analyses revealed that the AAVM41 capsid is a recombinant of AAV1, 6, 7, and 8 with a mosaic capsid surface and a conserved capsid interior. AAVM41 was then subjected to a side-by-side comparison to AAV9, the most robust AAV for systemic heart and muscle gene delivery; to AAV6, a parental AAV with strong muscle tropism. After i.v. delivery of reporter genes, AAVM41 was found more efficient than AAV6 in the heart and muscle, and was similar to AAV9 in the heart but weaker in the muscle. In fact, the myocardium showed the highest gene expression among all tissues tested in mice and hamsters after systemic AAVM41 delivery. However, gene transfer in non-muscle tissues, mainly the liver, was dramatically reduced. AAVM41 was further tested in a genetic cardiomyopathy hamster model and achieved efficient long-term delta-sarcoglycan gene expression and rescue of cardiac functions. Thus, direct in vivo panning of capsid libraries is a simple tool for the de-targeting and retargeting of viral vector tissue tropisms facilitated by acquisition of desirable sequences and properties.


Asunto(s)
Barajamiento de ADN , Dependovirus/genética , Corazón/virología , Miocardio/metabolismo , Selección Genética , Animales , Proteínas de la Cápside/química , Proteínas de la Cápside/genética , Cardiomiopatías/virología , Cricetinae , Modelos Animales de Enfermedad , Dosificación de Gen , Biblioteca de Genes , Vectores Genéticos/administración & dosificación , Genoma Viral/genética , Insuficiencia Cardíaca/virología , Humanos , Inmunoglobulina G/inmunología , Luciferasas/metabolismo , Ratones , Modelos Moleculares , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/virología , Pruebas de Neutralización , Especificidad de Órganos , Análisis de Secuencia de Proteína , Transducción Genética
13.
Mol Ther ; 18(8): 1501-8, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20517298

RESUMEN

Duchenne (DMD) and golden retriever (GRMD) muscular dystrophy are caused by genetic mutations in the dystrophin gene and afflict striated muscles. We investigated systemic gene delivery in 4-day-old GRMD dogs given a single intravenous injection of an AAV9 vector (1.5 x 10(14) vector genomes/kg) carrying a human codon-optimized human mini-dystrophin gene under control of the cytomegalovirus (CMV) promoter. One of the three treated dogs was euthanized 9 days later due to pre-existing conditions. Scattered mini-dystrophin-positive myofibers were seen by immunofluorescent (IF) staining in numerous muscles. At the end of the 16-week study, the other two dogs showed generalized muscle expression of mini-dystrophin in ~15% to nearly 100% of myofibers. Western blot and vector DNA quantitative PCR results agreed with the IF data. Delayed growth and pelvic limb muscle atrophy and contractures were seen several weeks after vector delivery. T-2 weighted magnetic resonance imaging (MRI) at 8 weeks showed increased signal intensity compatible with inflammation in several pelvic limb muscles. This marked early inflammatory response raised concerns regarding methodology. Use of the ubiquitous CMV promoter, extra-high vector dose, and marked expression of a human protein in canine muscles may have contributed to the pathologic changes seen in the pelvic limbs.


Asunto(s)
Adenoviridae/genética , Distrofina/deficiencia , Distrofina/metabolismo , Terapia Genética/métodos , Vectores Genéticos/genética , Inyecciones Intravenosas/métodos , Músculos/metabolismo , Animales , Animales Recién Nacidos , Western Blotting , Perros , Distrofina/genética , Femenino , Vectores Genéticos/administración & dosificación , Humanos , Imagen por Resonancia Magnética , Músculos/patología , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/metabolismo , Distrofia Muscular Animal/terapia
14.
Mol Ther Methods Clin Dev ; 22: 196-209, 2021 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-34485605

RESUMEN

Fragile X syndrome (FXS), a neurodevelopmental disorder with no known cure, is caused by a lack of expression of the fragile X mental retardation protein (FMRP). As a single-gene disorder, FXS is an excellent candidate for viral-vector-based gene therapy, although that is complicated by the existence of multiple isoforms of FMRP, whose individual cellular functions are unknown. We studied the effects of rat and mouse orthologs of human isoform 17, a major expressed isoform of FMRP. Injection of neonatal Fmr1 knockout rats and mice with adeno-associated viral vectors (AAV9 serotype) under the control of an MeCP2 mini-promoter resulted in widespread distribution of the FMRP transgenes throughout the telencephalon and diencephalon. Transgene expression occurred mainly in non-GABAergic neurons, with little expression in glia. Early postnatal treatment resulted in partial rescue of the Fmr1 KO rat phenotype, including improved social dominance in treated Fmr1 KO females and partial rescue of locomotor activity in males. Electro-encephalogram (EEG) recordings showed correction of abnormal slow-wave activity during the sleep-like state in male Fmr1 KO rats. These findings support the use of AAV-based gene therapy as a treatment for FXS and specifically demonstrate the potential therapeutic benefit of human FMRP isoform 17 orthologs.

15.
Skelet Muscle ; 9(1): 16, 2019 05 27.
Artículo en Inglés | MEDLINE | ID: mdl-31133057

RESUMEN

BACKGROUND: Growth differentiation factor 11 (GDF11) is a member of the transforming growth factor ß superfamily. The GDF11 propeptide, which is derived from the GDF11 precursor protein, blocks the activity of GDF11 and its homolog, myostatin, which are both potent inhibitors of muscle growth. Thus, treatment with GDF11 propeptide may be a potential therapeutic strategy for diseases associated with muscle atrophy like sarcopenia and the muscular dystrophies. Here, we evaluate the impact of GDF11 propeptide-Fc (GDF11PRO-Fc) gene delivery on skeletal muscle in normal and dystrophic adult mice. METHODS: A pull-down assay was used to obtain physical confirmation of a protein-protein interaction between GDF11PRO-Fc and GDF11 or myostatin. Next, differentiated C2C12 myotubes were treated with AAV6-GDF11PRO-Fc and challenged with GDF11 or myostatin to determine if GDF11PRO-Fc could block GDF11/myostatin-induced myotube atrophy. Localized expression of GDF11PRO-Fc was evaluated via a unilateral intramuscular injection of AAV9-GDF11PRO-Fc into the hindlimb of C57BL/6J mice. In mdx mice, intravenous injection of AAV9-GDF11PRO-Fc was used to achieve systemic expression. The impact of GDF11PRO-Fc on muscle mass, function, and pathological features were assessed. RESULTS: GDF11PRO-Fc was observed to bind both GDF11 and myostatin. In C2C12 myotubes, expression of GDF11PRO-Fc was able to mitigate GDF11/myostatin-induced atrophy. Following intramuscular injection in C57BL/6J mice, increased grip strength and localized muscle hypertrophy were observed in the injected hindlimb after 10 weeks. In mdx mice, systemic expression of GDF11PRO-Fc resulted in skeletal muscle hypertrophy without a significant change in cardiac mass after 12 weeks. In addition, grip strength and rotarod latency time were improved. Intramuscular fibrosis was also reduced in treated mdx mice; however, there was no change seen in central nucleation, membrane permeability to serum IgG or serum creatine kinase levels. CONCLUSIONS: GDF11PRO-Fc induces skeletal muscle hypertrophy and improvements in muscle strength via inhibition of GDF11/myostatin signaling. However, GDF11PRO-Fc does not significantly improve the dystrophic pathology in mdx mice.


Asunto(s)
Proteínas Morfogenéticas Óseas/antagonistas & inhibidores , Factores de Diferenciación de Crecimiento/antagonistas & inhibidores , Distrofia Muscular Animal/tratamiento farmacológico , Miostatina/antagonistas & inhibidores , Animales , Proteínas Morfogenéticas Óseas/genética , Proteínas Morfogenéticas Óseas/metabolismo , Terapia Genética , Vectores Genéticos , Factores de Diferenciación de Crecimiento/genética , Factores de Diferenciación de Crecimiento/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Fuerza Muscular/efectos de los fármacos , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/patología , Músculo Esquelético/fisiopatología , Distrofia Muscular Animal/patología , Distrofia Muscular Animal/fisiopatología , Distrofia Muscular de Duchenne/tratamiento farmacológico , Distrofia Muscular de Duchenne/patología , Distrofia Muscular de Duchenne/fisiopatología , Miostatina/metabolismo , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , Precursores de Proteínas/farmacología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes de Fusión/farmacología
16.
Hum Gene Ther ; 19(3): 241-54, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18288893

RESUMEN

Myostatin has been extensively documented as a negative regulator of muscle growth. Myostatin inhibition is therefore considered an attractive strategy for the treatment of muscle-wasting diseases such as muscular dystrophies. To investigate whether systemic gene delivery of myostatin propeptide (MRPO), a natural inhibitor of myostatin, could enhance body-wide skeletal muscle growth, we used adeno-associated virus serotype 8 (AAV8) vectors to deliver the MRPO gene into either normal mice or mdx mice, a murine model of Duchenne muscular dystrophy (DMD). In normal mice, a significant increase in skeletal muscle mass was observed after either an intraperitoneal injection of AAV-MPRO into neonates, or an intravenous injection of AAV-MPRO76AFc (a modified MPRO fused with IgG Fc) into adults. Enhanced muscle growth occurred because of myofiber hypertrophy, not hyperplasia. In mdx mice, a significant increase in skeletal muscle mass was also observed after AAV-MPRO76AFc injection. The treated mdx mice showed larger and more uniform myofibers, fewer infiltrating mononuclear cells, less fibrosis, and lower serum creatine kinase levels. In addition, a grip force test and an in vitro tetanic contractile force test showed improved muscle strength. A treadmill test, however, showed reduced endurance of the treated mdx mice compared with their untreated counterparts. Importantly, no cardiac hypertrophy was observed in either normal or mdx mice after myostatin inhibition by gene delivery. These results clearly demonstrate the efficacy of AAV8-mediated myostatin propeptide gene delivery in a rodent model of DMD, and warrant further investigation in large animal models and eventually in human patients.


Asunto(s)
Terapia Genética , Desarrollo de Músculos , Fibras Musculares Esqueléticas/fisiología , Músculo Esquelético/crecimiento & desarrollo , Distrofia Muscular Animal/terapia , Péptidos/genética , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Factor de Crecimiento Transformador beta/genética , Animales , Animales Recién Nacidos , Dependovirus/genética , Modelos Animales de Enfermedad , Vectores Genéticos , Ratones , Ratones Endogámicos mdx , Músculo Esquelético/citología , Distrofia Muscular Animal/genética , Miostatina , Fenotipo
17.
Nat Biotechnol ; 23(3): 321-8, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15735640

RESUMEN

Systemic gene delivery into muscle has been a major challenge for muscular dystrophy gene therapy, with capillary blood vessels posing the principle barrier and limiting vector dissemination. Previous efforts to deliver genes into multiple muscles have relied on isolated vessel perfusion or pharmacological interventions to enforce broad vector distribution. We compared the efficiency of multiple adeno-associated virus (AAV) vectors after a single injection via intraperitoneal or intravenous routes without additional intervention. We show that AAV8 is the most efficient vector for crossing the blood vessel barrier to attain systemic gene transfer in both skeletal and cardiac muscles of mice and hamsters. Serotypes such as AAV1 and AAV6, which demonstrate robust infection in skeletal muscle cells, were less effective in crossing the blood vessel barrier. Gene expression persisted in muscle and heart, but diminished in tissues undergoing rapid cell division, such as neonatal liver. This technology should prove useful for muscle-directed systemic gene therapy.


Asunto(s)
Dependovirus/genética , Sistemas de Liberación de Medicamentos/métodos , Marcación de Gen/métodos , Corazón/virología , Músculo Esquelético/metabolismo , Músculo Esquelético/virología , Miocardio/metabolismo , Transducción Genética/métodos , Animales , Animales Recién Nacidos , Cricetinae , ADN/administración & dosificación , Terapia Genética/métodos , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR
18.
Mol Ther Nucleic Acids ; 12: 283-293, 2018 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-30195767

RESUMEN

Duchenne muscular dystrophy (DMD) is a severe muscle-wasting disorder caused by a mutation in the dystrophin gene. Numerous gene therapies have been developed to replace or repair the defective dystrophin gene; however, these treatments cannot restore the full-length protein or completely resolve dystrophic symptoms. Secondary pathological mechanisms, such as functional ischemia and fibrosis, are thought to exacerbate the primary defect and cause the profound muscle degeneration found in dystrophic muscle. Surrogate therapies utilizing alternative therapeutic genes, or "booster genes," such as VEGFA and utrophin, seek to address these secondary mechanisms and have shown impressive benefit in mdx mice. A skeletal muscle-specific microRNA, miR-206, is particularly overexpressed in dystrophic muscle and inhibits the expression of known booster genes. Thus, we aimed to determine if miR-206 contributes to dystrophic pathology by repressing beneficial gene expression. Here, we show that AAV-mediated expression of a miR-206 decoy target effectively downregulated miR-206 expression and increased endogenous therapeutic gene expression in mature mdx muscle. Furthermore, treatment significantly improved motor function and dystrophic pathology in mdx mice. In summary, we have identified a contributing factor to the dystrophic phenotype and characterized a novel therapeutic avenue for DMD.

19.
Mol Ther Methods Clin Dev ; 9: 47-56, 2018 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-29766020

RESUMEN

LAMA2-related muscular dystrophy (LAMA2 MD) is the most common and fatal form of early-onset congenital muscular dystrophies. Due to the large size of the laminin α2 cDNA and heterotrimeric structure of the protein, it is challenging to develop a gene-replacement therapy. Our group has developed a novel adeno-associated viral (AAV) vector carrying the mini-agrin, which is a non-homologous functional substitute for the mutated laminin α2. A significant therapeutic effect in skeletal muscle was observed in our previous study using AAV serotype 1 (AAV1). In this investigation, we examined AAV9 vector, which has more widespread transduction than AAV1, to determine if the therapeutic effects could be further improved. As expected, AAV9-mini-agrin treatment offered enhanced therapeutic effects over the previously used AAV1-mini-agrin in extending mouse lifespan and improvement of muscle pathology. Additionally, overexpression of mini-agrin in peripheral nerves of dyw/dyw mice partially amended nerve pathology as evidenced by improved motor function and sensorimotor processing, partial restoration of myelination, partial restoration of basement membrane via EM examination, as well as decreased regeneration of Schwann cells. In conclusion, our studies indicate that overexpression of mini-agrin into dyw/dyw mice offers profound therapeutic effects in both skeletal muscle and nervous system.

20.
Exp Mol Med ; 49(9): e377, 2017 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-28912572

RESUMEN

Delivery of follistatin (FST) represents a promising strategy for both muscular dystrophies and diabetes, as FST is a robust antagonist of myostatin and activin, which are critical regulators of skeletal muscle and adipose tissues. FST is a multi-domain protein, and deciphering the function of different domains will facilitate novel designs for FST-based therapy. Our study aims to investigate the role of the N-terminal domain (ND) of FST in regulating muscle and fat mass in vivo. Different FST constructs were created and packaged into the adeno-associated viral vector (AAV). Overexpression of wild-type FST in normal mice greatly increased muscle mass while decreasing fat accumulation, whereas overexpression of an N terminus mutant or N terminus-deleted FST had no effect on muscle mass but moderately decreased fat mass. In contrast, FST-I-I containing the complete N terminus and double domain I without domain II and III had no effect on fat but increased skeletal muscle mass. The effects of different constructs on differentiated C2C12 myotubes were consistent with the in vivo finding. We hypothesized that ND was critical for myostatin blockade, mediating the increase in muscle mass, and was less pivotal for activin binding, which accounts for the decrease in the fat tissue. An in vitro TGF-beta1-responsive reporter assay revealed that FST-I-I and N terminus-mutated or -deleted FST showed differential responses to blockade of activin and myostatin. Our study provided direct in vivo evidence for a role of the ND of FST, shedding light on future potential molecular designs for FST-based gene therapy.


Asunto(s)
Tejido Adiposo/anatomía & histología , Tejido Adiposo/metabolismo , Folistatina/metabolismo , Músculo Esquelético/anatomía & histología , Músculo Esquelético/metabolismo , Dominios y Motivos de Interacción de Proteínas , Animales , Biomarcadores , Diferenciación Celular/genética , Línea Celular , Dependovirus/genética , Femenino , Técnica del Anticuerpo Fluorescente , Folistatina/química , Folistatina/genética , Expresión Génica , Orden Génico , Genes Reporteros , Vectores Genéticos/genética , Humanos , Ratones , Mutación , Mioblastos/citología , Mioblastos/metabolismo , Tamaño de los Órganos , Regiones Promotoras Genéticas , Dominios y Motivos de Interacción de Proteínas/genética , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA