Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Ther ; 32(8): 2604-2623, 2024 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-38910327

RESUMEN

Recent clinical studies of single gene replacement therapy for neuromuscular disorders have shown they can slow or stop disease progression, but such therapies have had little impact on reversing muscle disease that was already present. To reverse disease in patients with muscular dystrophy, new muscle mass and strength must be rebuilt at the same time that gene replacement prevents subsequent disease. Here, we show that treatment of FKRPP448L mice with a dual FKRP/FST gene therapy packaged into a single adeno-associated virus (AAV) vector can build muscle strength and mass that exceed levels found in wild-type mice and can induce normal ambulation endurance in a 1-h walk test. Dual FKRP/FST therapy also showed more even increases in muscle mass and amplified muscle expression of both genes relative to either single gene therapy alone. These data suggest that treatment with single AAV-bearing dual FKRP/FST gene therapies can overcome loss of ambulation by improving muscle strength at the same time it prevents subsequent muscle damage. This design platform could be used to create therapies for other forms of muscular dystrophy that may improve patient outcomes.


Asunto(s)
Dependovirus , Modelos Animales de Enfermedad , Terapia Genética , Vectores Genéticos , Fuerza Muscular , Músculo Esquelético , Pentosiltransferasa , Animales , Ratones , Terapia Genética/métodos , Fuerza Muscular/genética , Dependovirus/genética , Vectores Genéticos/genética , Vectores Genéticos/administración & dosificación , Pentosiltransferasa/genética , Pentosiltransferasa/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Expresión Génica , Caminata , Humanos , Regulación de la Expresión Génica
2.
Am J Pathol ; 191(8): 1474-1486, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34294193

RESUMEN

Humans cannot synthesize the common mammalian sialic acid N-glycolylneuraminic acid (Neu5Gc) because of an inactivating deletion in the cytidine-5'-monophospho-(CMP)-N-acetylneuraminic acid hydroxylase (CMAH) gene responsible for its synthesis. Human Neu5Gc deficiency can lead to development of anti-Neu5Gc serum antibodies, the levels of which can be affected by Neu5Gc-containing diets and by disease. Metabolic incorporation of dietary Neu5Gc into human tissues in the face of circulating antibodies against Neu5Gc-bearing glycans is thought to exacerbate inflammation-driven diseases like cancer and atherosclerosis. Probing of sera with sialoglycan arrays indicated that patients with Duchenne muscular dystrophy (DMD) had a threefold increase in overall anti-Neu5Gc antibody titer compared with age-matched controls. These antibodies recognized a broad spectrum of Neu5Gc-containing glycans. Human-like inactivation of the Cmah gene in mice is known to modulate severity in a variety of mouse models of human disease, including the X chromosome-linked muscular dystrophy (mdx) model for DMD. Cmah-/-mdx mice can be induced to develop anti-Neu5Gc-glycan antibodies as humans do. The presence of anti-Neu5Gc antibodies, in concert with induced Neu5Gc expression, correlated with increased severity of disease pathology in Cmah-/-mdx mice, including increased muscle fibrosis, expression of inflammatory markers in the heart, and decreased survival. These studies suggest that patients with DMD who harbor anti-Neu5Gc serum antibodies might exacerbate disease severity when they ingest Neu5Gc-rich foods, like red meats.


Asunto(s)
Autoanticuerpos/sangre , Distrofia Muscular de Duchenne/inmunología , Distrofia Muscular de Duchenne/patología , Ácidos Neuramínicos/sangre , Ácidos Neuramínicos/inmunología , Animales , Autoanticuerpos/inmunología , Autoantígenos/inmunología , Niño , Modelos Animales de Enfermedad , Humanos , Masculino , Ratones , Ratones Endogámicos mdx , Ratones Noqueados , Distrofia Muscular de Duchenne/sangre
3.
Mol Ther ; 27(3): 636-649, 2019 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-30711447

RESUMEN

Dilated cardiomyopathy is a common cause of death in patients with Duchenne muscular dystrophy (DMD). Gene therapies for DMD must, therefore, have a therapeutic impact in cardiac as well as skeletal muscles. Our previous studies have shown that GALGT2 overexpression in mdx skeletal muscles can prevent muscle damage. Here we have tested whether rAAVrh74.MCK.GALGT2 gene therapy in mdx cardiac muscle can prevent the loss of heart function. Treatment of mdx hearts with rAAVrh74.MCK.GALGT2 1 day after birth did not negatively alter hemodynamic function, tested at 3 months of age, and it prevented early left ventricular remodeling and expression of fibrotic gene markers. Intravenous treatment of mdx mice with rAAVrh74.MCK.GALGT2 at 2 months of age significantly improved stroke volume and cardiac output compared to mock-treated mice analyzed at 17 months, both at rest and after stimulation with dobutamine. rAAVrh74.MCK.GALGT2 treatment of mdx heart correlated with increased glycosylation of α-dystroglycan with the CT glycan and increased utrophin protein expression. These data provide the first demonstration that GALGT2 overexpression can inhibit the loss of cardiac function in the dystrophin-deficient heart and, thus, may benefit both cardiac and skeletal muscles in DMD patients.


Asunto(s)
Glicosiltransferasas/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/patología , Animales , Modelos Animales de Enfermedad , Distrofina/metabolismo , Terapia Genética , Glicosiltransferasas/genética , Corazón/fisiología , Immunoblotting , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Distrofia Muscular de Duchenne/terapia , Utrofina/metabolismo
4.
J Immunol ; 198(6): 2366-2373, 2017 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-28148732

RESUMEN

Humans and chimpanzees are more sensitive to endotoxin than are mice or monkeys, but any underlying differences in inflammatory physiology have not been fully described or understood. We studied innate immune responses in Cmah-/- mice, emulating human loss of the gene encoding production of Neu5Gc, a major cell surface sialic acid. CMP-N-acetylneuraminic acid hydroxylase (CMAH) loss occurred ∼2-3 million years ago, after the common ancestor of humans and chimpanzees, perhaps contributing to speciation of the genus HomoCmah-/- mice manifested a decreased survival in endotoxemia following bacterial LPS injection. Macrophages from Cmah-/- mice secreted more inflammatory cytokines with LPS stimulation and showed more phagocytic activity. Macrophages and whole blood from Cmah-/- mice also killed bacteria more effectively. Metabolic reintroduction of Neu5Gc into Cmah-/- macrophages suppressed these differences. Cmah-/- mice also showed enhanced bacterial clearance during sublethal lung infection. Although monocytes and monocyte-derived macrophages from humans and chimpanzees exhibited marginal differences in LPS responses, human monocyte-derived macrophages killed Escherichia coli and ingested E. coli BioParticles better. Metabolic reintroduction of Neu5Gc into human macrophages suppressed these differences. Although multiple mechanisms are likely involved, one cause is altered expression of C/EBPß, a transcription factor affecting macrophage function. Loss of Neu5Gc in Homo likely had complex effects on immunity, providing greater capabilities to clear sublethal bacterial challenges, possibly at the cost of endotoxic shock risk. This trade-off may have provided a selective advantage when Homo transitioned to butchery using stone tools. The findings may also explain why the Cmah-/- state alters severity in mouse models of human disease.


Asunto(s)
Endotoxemia/inmunología , Escherichia coli/fisiología , Inflamación/inmunología , Macrófagos/inmunología , Oxigenasas de Función Mixta/metabolismo , Animales , Bacteriólisis/genética , Evolución Biológica , Diferenciación Celular , Linaje de la Célula , Células Cultivadas , Femenino , Humanos , Lipopolisacáridos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Oxigenasas de Función Mixta/genética , Pan troglodytes , Fagocitosis/genética
5.
Am J Pathol ; 186(9): 2429-48, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27561302

RESUMEN

Overexpression of B4GALNT2 (previously GALGT2) inhibits the development of muscle pathology in mouse models of Duchenne muscular dystrophy, congenital muscular dystrophy 1A, and limb girdle muscular dystrophy 2D. In these models, muscle GALGT2 overexpression induces the glycosylation of α dystroglycan with the cytotoxic T cell glycan and increases the overexpression of dystrophin and laminin α2 surrogates known to inhibit disease. Here, we show that GALGT2 gene therapy significantly reduces muscle pathology in FKRP P448Lneo(-) mice, a model for limb girdle muscular dystrophy 2I. rAAVrh74.MCK.GALGT2-treated FKRP P448Lneo(-) muscles showed reduced levels of centrally nucleated myofibers, reduced variance, increased size of myofiber diameters, reduced myofiber immunoglobulin G uptake, and reduced muscle wasting at 3 and 6 months after treatment. GALGT2 overexpression in FKRP P448Lneo(-) muscles did not cause substantial glycosylation of α dystroglycan with the cytotoxic T cell glycan or increased expression of dystrophin and laminin α2 surrogates in mature skeletal myofibers, but it increased the number of embryonic myosin-positive regenerating myofibers. These data demonstrate that GALGT2 overexpression can reduce the extent of muscle pathology in FKRP mutant muscles, but that it may do so via a mechanism that differs from its ability to induce surrogate gene expression.


Asunto(s)
Terapia Genética/métodos , Glicosiltransferasas/genética , Músculo Esquelético/patología , Distrofia Muscular de Cinturas/patología , Distrofia Muscular Animal/patología , Animales , Immunoblotting , Inmunohistoquímica , Inmunoprecipitación , Ratones , Ratones Mutantes , Mutación , Pentosiltransferasa , Proteínas/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Transferasas
6.
Am J Pathol ; 185(10): 2668-84, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26435413

RESUMEN

Transgenic overexpression of Galgt2 (official name B4Galnt2) in skeletal muscle stimulates the glycosylation of α dystroglycan (αDG) and the up-regulation of laminin α2 and dystrophin surrogates known to inhibit muscle pathology in mouse models of congenital muscular dystrophy 1A and Duchenne muscular dystrophy. Skeletal muscle Galgt2 gene expression is also normally increased in the mdx mouse model of Duchenne muscular dystrophy compared with the wild-type mice. To assess whether this increased endogenous Galgt2 expression could affect disease, we quantified muscular dystrophy measures in mdx mice deleted for Galgt2 (Galgt2(-/-)mdx). Galgt2(-/-) mdx mice had increased heart and skeletal muscle pathology and inflammation, and also worsened cardiac function, relative to age-matched mdx mice. Deletion of Galgt2 in wild-type mice also slowed skeletal muscle growth in response to acute muscle injury. In each instance where Galgt2 expression was elevated (developing muscle, regenerating muscle, and dystrophic muscle), Galgt2-dependent glycosylation of αDG was also increased. Overexpression of Galgt2 failed to inhibit skeletal muscle pathology in dystroglycan-deficient muscles, in contrast to previous studies in dystrophin-deficient mdx muscles. This study demonstrates that Galgt2 gene expression and glycosylation of αDG are dynamically regulated in muscle and that endogenous Galgt2 gene expression can ameliorate the extent of muscle pathology, inflammation, and dysfunction in mdx mice.


Asunto(s)
Distrofina/metabolismo , Glicosiltransferasas/metabolismo , Músculo Esquelético/patología , Distrofia Muscular de Duchenne/metabolismo , Animales , Modelos Animales de Enfermedad , Distrofina/deficiencia , Glicosiltransferasas/genética , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Ratones Endogámicos mdx , Ratones Noqueados , Desarrollo de Músculos/genética , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/patología , Miositis/patología , Regulación hacia Arriba
7.
Mol Ther ; 22(4): 713-24, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24145553

RESUMEN

Overexpression of GALGT2 in skeletal muscle can stimulate the glycosylation of α dystroglycan and the upregulation of normally synaptic dystroglycan-binding proteins, some of which are dystrophin and laminin α2 surrogates known to be therapeutic for several forms of muscular dystrophy. This article describes the vascular delivery of GALGT2 gene therapy in a large animal model, the rhesus macaque. Recombinant adeno-associated virus, rhesus serotype 74 (rAAVrh74), was used to deliver GALGT2 via the femoral artery to the gastrocnemius muscle using an isolated focal limb perfusion method. GALGT2 expression averaged 44 ± 4% of myofibers after treatment in macaques with low preexisting anti-rAAVrh74 serum antibodies, and expression was reduced to 9 ± 4% of myofibers in macaques with high preexisting rAAVrh74 immunity (P < 0.001; n = 12 per group). This was the case regardless of the addition of immunosuppressants, including prednisolone, tacrolimus, and mycophenolate mofetil. GALGT2-treated macaque muscles showed increased glycosylation of α dystroglycan and increased expression of dystrophin and laminin α2 surrogate proteins, including utrophin, plectin1, agrin, and laminin α5. These experiments demonstrate successful transduction of rhesus macaque muscle with rAAVrh74.MCK.GALGT2 after vascular delivery and induction of molecular changes thought to be therapeutic in several forms of muscular dystrophy.


Asunto(s)
Distrofina/biosíntesis , Técnicas de Transferencia de Gen , Terapia Genética , Laminina/biosíntesis , Distrofias Musculares/genética , Animales , Dependovirus/genética , Modelos Animales de Enfermedad , Distroglicanos/genética , Distroglicanos/metabolismo , Distrofina/genética , Regulación de la Expresión Génica , Glicosiltransferasas/genética , Laminina/genética , Macaca mulatta/genética , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Distrofias Musculares/patología , Distrofias Musculares/terapia
8.
Neurobiol Dis ; 62: 31-43, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24021662

RESUMEN

Recent clinical and pre-clinical studies suggest that both active and passive immunization strategies targeting Aß amyloid may have clinical benefit in Alzheimer's disease. Here, we demonstrate that vaccination of APPswePSEN1dE9 mice with SDPM1, an engineered non-native Aß amyloid-specific binding peptide, lowers brain Aß amyloid plaque burden and brain Aß1-40 and Aß1-42 peptide levels, improves cognitive learning and memory in Morris water maze tests and increases the expression of synaptic brain proteins. This was the case in young mice immunized prior to development of significant brain amyloid burden, and in older mice, where brain amyloid was already present. Active immunization was optimized using ALUM as an adjuvant to stimulate production of anti-SDPM1 and anti-Aß amyloid antibodies. Intracerebral injection of P4D6, an SDPM1 peptide-mimotope antibody, also lowered brain amyloid plaque burden in APPswePSEN1dE9 mice. Additionally, P4D6 inhibited Aß amyloid-mediated toxicity in cultured neuronal cells. The protein sequence of the variable domain within the P4D6 heavy chain was found to mimic a multimer of the SDPM1 peptide motif. These data demonstrate the efficacy of active and passive vaccine strategies to target Aß amyloid oligomers using an engineered peptide-mimotope strategy.


Asunto(s)
Enfermedad de Alzheimer/terapia , Vacunas contra el Alzheimer/uso terapéutico , Péptidos/uso terapéutico , Hidróxido de Aluminio/inmunología , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/prevención & control , Péptidos beta-Amiloides/metabolismo , Animales , Corteza Cerebral/patología , Modelos Animales de Enfermedad , Hipocampo/patología , Inmunización Pasiva , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Ratones Transgénicos , Fragmentos de Péptidos/metabolismo , Placa Amiloide/patología , Sinapsis/metabolismo , Resultado del Tratamiento , Vacunación
9.
Glycobiology ; 23(7): 833-43, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23514716

RESUMEN

Roughly 3 million years ago, an inactivating deletion occurred in CMAH, the human gene encoding CMP-Neu5Ac (cytidine-5'-monophospho-N-acetylneuraminic acid) hydroxylase (Chou HH, Takematsu H, Diaz S, Iber J, Nickerson E, Wright KL, Muchmore EA, Nelson DL, Warren ST, Varki A. 1998. A mutation in human CMP-sialic acid hydroxylase occurred after the Homo-Pan divergence. Proc Natl Acad Sci USA. 95:11751-11756). This inactivating deletion is now homozygous in all humans, causing the loss of N-glycolylneuraminic acid (Neu5Gc) biosynthesis in all human cells and tissues. The CMAH enzyme is active in other mammals, including mice, where Neu5Gc is an abundant form of sialic acid on cellular membranes, including those in cardiac and skeletal muscle. We recently demonstrated that the deletion of mouse Cmah worsened the severity of pathophysiology measures related to muscular dystrophy in mdx mice, a model for Duchenne muscular dystrophy (Chandrasekharan K, Yoon JH, Xu Y, deVries S, Camboni M, Janssen PM, Varki A, Martin PT. 2010. A human-specific deletion in mouse Cmah increases disease severity in the mdx model of Duchenne muscular dystrophy. Sci Transl Med. 2:42-54). Here, we demonstrate similar changes in cardiac and skeletal muscle pathology and physiology resulting from Cmah deletion in α-sarcoglycan-deficient (Sgca(-/-)) mice, a model for limb girdle muscular dystrophy 2D. These experiments demonstrate that loss of mouse Cmah can worsen disease severity in more than one form of muscular dystrophy and suggest that Cmah may be a general genetic modifier of muscle disease.


Asunto(s)
Músculo Esquelético/patología , Miocardio/patología , Ácidos Neuramínicos/metabolismo , Sarcoglicanos/genética , Animales , Eliminación de Gen , Humanos , Ratones , Ratones Endogámicos C57BL , Músculo Esquelético/metabolismo , Músculo Esquelético/fisiopatología , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/metabolismo , Miocardio/metabolismo , Sarcoglicanos/deficiencia
10.
J Pathol ; 226(1): 40-9, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21915858

RESUMEN

We have previously reported that mice with muscular dystrophy, including mdx mice, develop embryonal rhabdomyosarcoma (eRMS) with a low incidence after 1 year of age and that almost all such tumours contain cancer-associated p53 mutations. To further demonstrate the relevance of p53 inactivation, we created p53-deficient mdx mice. Here we demonstrate that loss of one or both p53 (Trp53) alleles accelerates eRMS incidence in the mdx background, such that almost all Trp53(-/-) mdx animals develop eRMS by 5 months of age. To ascertain whether increased tumour incidence was due to the regenerative microenvironment found in dystrophic skeletal muscles, we induced muscle regeneration in Trp53(+/+) and Trp53(-/-) animals using cardiotoxin (Ctx). Wild-type (Trp53(+/+) ) animals treated with Ctx, either once every 7 days or once every 14 days from 1 month of age onwards, developed no eRMS; however, all similarly Ctx-treated Trp53(-/-) animals developed eRMS by 5 months of age at the site of injection. Most of these tumours displayed markers of human eRMS, including over-expression of Igf2 and phosphorylated Akt. These data demonstrate that the presence of a regenerative microenvironment in skeletal muscle, coupled with Trp53 deficiency, is sufficient to robustly induce eRMS in young mice. These studies further suggest that consideration should be given to the potential of the muscle microenvironment to support tumourigenesis in regenerative therapies for myopathies.


Asunto(s)
Músculo Esquelético/patología , Rabdomiosarcoma Embrionario/genética , Rabdomiosarcoma Embrionario/patología , Microambiente Tumoral/fisiología , Proteína p53 Supresora de Tumor/genética , Animales , Ratones , Ratones Endogámicos mdx , Ratones Noqueados , Músculo Esquelético/fisiología , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/patología , Regeneración
11.
Mol Cell Neurosci ; 51(3-4): 112-26, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22982027

RESUMEN

At the mammalian neuromuscular junction (NMJ), the CT (cytotoxic T cell) carbohydrate antigen [GalNAcß1,4[Neu5Ac/Gcα2,3]Galß1,4GlcNAc-] is a unique synaptic cell surface carbohydrate present in both the presynaptic and postsynaptic membranes. Here we show that Galgt1, which synthesizes the ß1,4GalNAc linkage of the CT carbohydrate on gangliosides, is required for presynaptic expression of the CT carbohydrate at the NMJ, while Galgt2, which can synthesize the ß1,4GalNAc of the CT carbohydrate on glycoproteins, is required for postsynaptic expression. Proper postsynaptic localization of the CT carbohydrate also required muscle expression of dystroglycan, a known muscle substrate for Galgt2. Transgenic overexpression of Galgt2 in skeletal myofibers altered the expression of synaptic muscle proteins and altered neuromuscular topography, which was partially NCAM-dependent, while an increase in postsynaptic AChR-rich domains was observed in both neuron- and skeletal muscle-specific Galgt2 transgenic mice. By contrast, overexpression of Galgt1 in muscle did not allow for increased expression of CT carbohydrate on the sarcolemmal membrane and instead caused muscle pathology. Loss of Galgt2 increased intracellular accumulation of acetylcholine receptors and acetylcholinesterase within skeletal myofibers, suggesting an additional role for Galgt2 in neuromuscular stability. These experiments demonstrate that Galgt1 and Galgt2 contribute in distinct ways to the expression and function of synaptic ßGalNAc-containing carbohydrates at the NMJ.


Asunto(s)
Antígenos de Grupos Sanguíneos/metabolismo , Glicosiltransferasas/metabolismo , Unión Neuromuscular/metabolismo , Acetilcolinesterasa/metabolismo , Acetilgalactosamina/metabolismo , Acetilglucosamina/metabolismo , Animales , Antígenos de Superficie/química , Antígenos de Superficie/metabolismo , Antígenos de Grupos Sanguíneos/análisis , Antígenos de Grupos Sanguíneos/inmunología , Distroglicanos/metabolismo , Gangliósidos/metabolismo , Glicosiltransferasas/genética , Ratones , Ratones Transgénicos , Fibras Musculares Esqueléticas/enzimología , Proteínas Musculares/metabolismo , Unión Neuromuscular/enzimología , Receptores Colinérgicos/metabolismo , Sarcolema/metabolismo
12.
J Neuromuscul Dis ; 10(5): 797-812, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37458043

RESUMEN

BACKGROUND: GNE myopathy (GNEM) is a severe muscle disease caused by mutations in the UDP-GlcNAc-2-epimerase/ManNAc-6-kinase (GNE) gene, which encodes a bifunctional enzyme required for sialic acid (Sia) biosynthesis. OBJECTIVE: To develop assays to demonstrate the potency of AAV gene therapy vectors in making Sia and to define the dose required for replacement of endogenous mouse Gne gene expression with human GNE in skeletal muscles. METHODS: A MyoD-inducible Gne-deficient cell line, Lec3MyoDI, and a GNE-deficient human muscle cell line, were made and tested to define the potency of various AAV vectors to increase binding of Sia-specific lectins, including MAA and SNA. qPCR and qRT-PCR methods were used to quantify AAV biodistribution and GNE gene expression after intravenous delivery of AAV vectors designed with different promoters in wild-type mice. RESULTS: Lec3 cells showed a strong deficit in MAA binding, while GNE-/-MB135 cells did not. Overexpressing GNE in Lec3 and Lec3MyoDI cells by AAV infection stimulated MAA binding in a dose-dependent manner. Use of a constitutive promoter, CMV, showed higher induction of MAA binding than use of muscle-specific promoters (MCK, MHCK7). rAAVrh74.CMV.GNE stimulated human GNE expression in muscles at levels equivalent to endogenous mouse Gne at a dose of 1×1013vg/kg, while AAVs with muscle-specific promoters required higher doses. AAV biodistribution in skeletal muscles trended higher when CMV was used as the promoter, and this correlated with increased sialylation of its viral capsid. CONCLUSIONS: Lec3 and Lec3MyoDI cells work well to assay the potency of AAV vectors in making Sia. Systemic delivery of rAAVrh74.CMV.GNE can deliver GNE gene replacement to skeletal muscles at doses that do not overwhelm non-muscle tissues, suggesting that AAV vectors that drive constitutive organ expression could be used to treat GNEM.


Asunto(s)
Infecciones por Citomegalovirus , Músculo Esquelético , Humanos , Ratones , Animales , Distribución Tisular , Músculo Esquelético/metabolismo , Ácido N-Acetilneuramínico/metabolismo , Terapia Genética , Infecciones por Citomegalovirus/metabolismo
13.
Eur J Hum Genet ; 31(6): 663-673, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36935420

RESUMEN

The major determinant of disease severity in Duchenne muscular dystrophy (DMD) or milder Becker muscular dystrophy (BMD) is whether the dystrophin gene (DMD) mutation truncates the mRNA reading frame or allows expression of a partially functional protein. However, even in the complete absence of dystrophin, variability in disease severity is observed, and candidate gene studies have implicated several genes as modifiers. Here we present the largest genome-wide search to date for loci influencing severity in N = 419 DMD patients. Availability of subjects for such studies is quite limited, leading to modest sample sizes, which present a challenge for GWAS design. We have therefore taken special steps to minimize heterogeneity within our dataset at the DMD locus itself, taking a novel approach to mutation classification to effectively exclude the possibility of residual dystrophin expression, and utilized statistical methods that are well adapted to smaller sample sizes, including the use of a novel linear regression-like residual for time to ambulatory loss and the application of evidential statistics for the GWAS approach. Finally, we applied an unbiased in silico pipeline, utilizing functional genomic datasets to explore the potential impact of the best supported SNPs. In all, we obtained eight SNPs (out of 1,385,356 total) with posterior probability of trait-marker association (PPLD) ≥ 0.4, representing six distinct loci. Our analysis prioritized likely non-coding SNP regulatory effects on six genes (ETAA1, PARD6G, GALNTL6, MAN1A1, ADAMTS19, and NCALD), each with plausibility as a DMD modifier. These results support both recurrent and potentially new pathways for intervention in the dystrophinopathies.


Asunto(s)
Distrofina , Distrofia Muscular de Duchenne , Humanos , Distrofina/genética , Distrofina/metabolismo , Estudio de Asociación del Genoma Completo , Exones , Distrofia Muscular de Duchenne/diagnóstico , Distrofia Muscular de Duchenne/genética , Gravedad del Paciente , Caminata , Antígenos de Superficie
14.
J Proteome Res ; 11(9): 4413-24, 2012 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-22775139

RESUMEN

Dystroglycan is a major cell surface glycoprotein receptor for the extracellular matrix in skeletal muscle. Defects in dystroglycan glycosylation cause muscular dystrophy and alterations in dystroglycan glycosylation can impact extracellular matrix binding. Here we describe an immunoprecipitation technique that allows isolation of beta dystroglycan with members of the dystrophin-associated protein complex (DAPC) from detergent-solubilized skeletal muscle. Immunoprecipitation, coupled with shotgun proteomics, has allowed us to identify new dystroglycan-associated proteins and define changed associations that occur within the DAPC in dystrophic skeletal muscles. In addition, we describe changes that result from overexpression of Galgt2, a normally synaptic muscle glycosyltransferase that can modify alpha dystroglycan and inhibit the development of muscular dystrophy when it is overexpressed. These studies identify new dystroglycan-associated proteins that may participate in dystroglycan's roles, both positive and negative, in muscular dystrophy.


Asunto(s)
Distroglicanos/metabolismo , Distrofina/genética , Glicosiltransferasas/genética , Músculo Esquelético/química , Proteoma/análisis , Animales , Western Blotting , Distroglicanos/química , Distrofina/metabolismo , Complejo de Proteínas Asociado a la Distrofina/química , Complejo de Proteínas Asociado a la Distrofina/metabolismo , Glicosiltransferasas/metabolismo , Inmunoprecipitación , Interleucina-15/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Proteínas de Microfilamentos/metabolismo , Músculo Esquelético/metabolismo , Distrofias Musculares/genética , Distrofias Musculares/metabolismo , Mapeo de Interacción de Proteínas , Proteoma/química , Proteoma/metabolismo , Proteómica/métodos , Reproducibilidad de los Resultados
15.
Mol Cell Neurosci ; 48(1): 9-19, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21699983

RESUMEN

Cell surface carbohydrates define subpopulations of primary olfactory neurons whose axons terminate in select glomeruli in the olfactory bulb. The combination of carbohydrates present on axon subpopulations has been proposed to confer a unique identity that contributes to the establishment of the olfactory topographic map. We have identified a novel subpopulation of primary olfactory neurons in mice that express blood group carbohydrates with GalNAc-ß1,4[NeuAcα 2,3]Galß1 residues recognised by the CT1 antibody. The CT1 carbohydrate has been shown to modulate adhesion of nerve terminals to the extracellular matrix and to synaptic proteins. The axons of the CT1-positive primary olfactory neurons terminate in a subpopulation of glomeruli in the olfactory bulb. Four lines of evidence support the view that CT1 glomeruli are topographically fixed. First, CT1 glomeruli were restricted predominantly to the dorsomedial olfactory bulb and were absent from large patches of the ventrolateral bulb. Second, similar distributions were observed for CT1 glomeruli on both the left and right olfactory bulbs of each animal, and between animals. Third, CT1 glomeruli were typically present as small clusters of 2-4 glomeruli. Fourth, a single CT1 glomerulus was always apposed to the glomeruli innervated by axons expressing the M72 odorant receptor. We also show that the CT1 carbohydrate is lost in gain-of-function transgenic mice over-expressing the blood group A glycosyltransferase in which there is aberrant targeting of M72 axons. Taken together, these results suggest that the CT1 carbohydrate, together with other carbohydrates, contributes to axon guidance during the establishment of the olfactory topographic map.


Asunto(s)
Glucolípidos/química , Glucolípidos/metabolismo , Bulbo Olfatorio/anatomía & histología , Bulbo Olfatorio/metabolismo , Animales , Ratones , Ratones Transgénicos , Bulbo Olfatorio/crecimiento & desarrollo , Neuronas Receptoras Olfatorias/fisiología , Neuronas Receptoras Olfatorias/ultraestructura
16.
Mol Ther Methods Clin Dev ; 26: 413-426, 2022 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-36092360

RESUMEN

Lysosomal acid lipase deficiency (LAL-D) presents as one of two rare autosomal recessive diseases: Wolman disease (WD), a severe disorder presenting in infancy characterized by absent or very low LAL activity, and cholesteryl ester storage disease (CESD), a less severe, later onset disease form. Recent clinical studies have shown efficacy of enzyme replacement therapy for both forms of LAL-D; however, no gene therapy approach has yet been developed for clinical use. Here, we show that rscAAVrh74.miniCMV.LIPA gene therapy can significantly improve disease symptoms in the Lipa -/- mouse model of LAL-D. Treatment dramatically lowered hepatosplenomegaly, liver and spleen triglyceride and cholesterol levels, and serum expression of markers of liver damage. Measures of liver inflammation and fibrosis were also reduced. Treatment of young adult mice was more effective than treatment of neonates, and enzyme activity was elevated in serum, consistent with possible bystander effects. These results demonstrate that adeno associated virus (AAV)-mediated LIPA gene-replacement therapy may be a viable option to treat patients with LAL-D, particularly patients with CESD.

17.
J Neuromuscul Dis ; 9(1): 53-71, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34511508

RESUMEN

BACKGROUND: GNE myopathy (GNEM) is a rare, adult-onset, inclusion body myopathy that results from mutations in the GNE gene. GNE encodes UDP-GlcNAc epimerase/ManNAc-6 kinase, a protein with two enzymatic activities that comprise the committed step in biosynthesis of sialic acid (SA), an essential glycan that appears on the terminal positions of many extracellular oligosaccharide chains. These GNE mutations can cause a reduction of SA in many tissues, although pathology is restricted to skeletal muscles through a poorly understood mechanism. OBJECTIVE: Despite recent advances in the field, it remains unclear which therapeutic avenue is most promising for the restoration of SA level in skeletal muscle affected by GNEM. Our objective was to assess dietary and gene therapy strategies for GNEM in Cmah-deficient GNED207VTgGne-/- mice, a model that allows for the visualization of orally delivered N-glycolylneuraminic acid (Neu5Gc), one of the two predominant SA forms in muscle. METHODS: Methods included in situ physiology studies of the tibialis anterior muscle, studies of ambulation and limb grip strength, and muscle staining using MAA, SNA, and anti-Neu5Gc antibody, along with qPCR, qRT-PCR, western blot, and HPLC studies to assess virally introduced DNA, GNE gene expression, GNE protein expression, and SA expression. RESULTS: We found that a diet enriched in Neu5Gc-containing glycoproteins had no impact on Neu5Gc immunostaining in muscles of GNEM model mice. Delivery of a single high dose oral Neu5Gc therapy, however, did increase Neu5Gc immunostaining, though to levels below those found in wild type mice. Delivery of a single dose of GNE gene therapy using a recombinant Adeno Associated Virus (rAAV) vector with a liver-specific or a muscle-specific promoter both caused increased muscle Neu5Gc immunostaining that exceeded that seen with single dose monosaccharide therapy. CONCLUSIONS: Our findings indicate that dietary loading of Neu5Gc-containing glycoproteins is not effective in increasing muscle Neu5Gc expression, while single dose oral Neu5Gc monosaccharide or GNE gene therapy are. Neu5Gc immunostaining, however, showed greater changes than did lectin staining or HPLC analysis. Taken together, these results suggest that Neu5Gc immunostaining may be more sensitive technique to follow SA expression than other more commonly used methods and that liver expression of GNE may contribute overall muscle SA content.


Asunto(s)
Dietoterapia , Miopatías Distales/terapia , Terapia Genética , Complejos Multienzimáticos/genética , Músculo Esquelético/metabolismo , Ácido N-Acetilneuramínico/metabolismo , Animales , Modelos Animales de Enfermedad , Miopatías Distales/genética , Miopatías Distales/metabolismo , Humanos , Ratones , Ratones Transgénicos
18.
Mol Ther Methods Clin Dev ; 27: 47-60, 2022 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-36186954

RESUMEN

In a phase 1/2, open-label dose escalation trial, we delivered rAAVrh74.MCK.GALGT2 (also B4GALNT2) bilaterally to the legs of two boys with Duchenne muscular dystrophy using intravascular limb infusion. Subject 1 (age 8.9 years at dosing) received 2.5 × 1013 vector genome (vg)/kg per leg (5 × 1013 vg/kg total) and subject 2 (age 6.9 years at dosing) received 5 × 1013 vg/kg per leg (1 × 1014 vg/kg total). No serious adverse events were observed. Muscle biopsy evaluated 3 or 4 months post treatment versus baseline showed evidence of GALGT2 gene expression and GALGT2-induced muscle cell glycosylation. Functionally, subject 1 showed a decline in 6-min walk test (6MWT) distance; an increase in time to run 100 m, and a decline in North Star Ambulatory Assessment (NSAA) score until ambulation was lost at 24 months. Subject 2, treated at a younger age and at a higher dose, demonstrated an improvement over 24 months in NSAA score (from 20 to 23 points), an increase in 6MWT distance (from 405 to 478 m), and only a minimal increase in 100 m time (45.6-48.4 s). These data suggest preliminary safety at a dose of 1 × 1014 vg/kg and functional stabilization in one patient.

19.
Am J Pathol ; 176(1): 416-34, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20019182

RESUMEN

Altered expression of proteins in the dystrophin-associated glycoprotein complex results in muscular dystrophy and has more recently been implicated in a number of forms of cancer. Here we show that loss of either of two members of this complex, dystrophin in mdx mice or alpha sarcoglycan in Sgca(-/-) mice, results in the spontaneous development of muscle-derived embryonal rhabdomyosarcoma (RMS) after 1 year of age. Many mdx and Sgca(-/-) tumors showed increased expression of insulin-like growth factor 2, retinoblastoma protein, and phosphorylated Akt and decreased expression of phosphatase and tensin homolog gene, much as is found in a human RMS. Further, all mdx and Sgca(-/-) RMS analyzed had increased expression of p53 and murine double minute (mdm)2 protein and contained missense p53 mutations previously identified in human cancers. The mdx RMS also contained missense mutations in Mdm2 or alternatively spliced Mdm2 transcripts that lacked an exon encoding a portion of the p53-binding domain. No Pax3:Fkhr or Pax7:Fkhr translocation mRNA products were evident in any tumor. Expression of natively glycosylated alpha dystroglycan and alpha sarcoglycan was reduced in mdx RMS, whereas dystrophin expression was absent in almost all human RMS, both for embryonal and alveolar RMS subtypes. These studies show that absence of members of the dystrophin-associated glycoprotein complex constitutes a permissive environment for spontaneous development of embryonal RMS associated with mutation of p53 and mutation or altered splicing of Mdm2.


Asunto(s)
Empalme Alternativo/genética , Distrofina/deficiencia , Mutación/genética , Proteínas Proto-Oncogénicas c-mdm2/genética , Rabdomiosarcoma Embrionario/patología , Sarcoglicanos/deficiencia , Proteína p53 Supresora de Tumor/genética , Envejecimiento/patología , Animales , Muerte Celular , Distrofina/metabolismo , Regulación Neoplásica de la Expresión Génica , Glicosilación , Humanos , Ratones , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Lesiones Precancerosas/genética , Lesiones Precancerosas/patología , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Rabdomiosarcoma Alveolar/genética , Rabdomiosarcoma Alveolar/patología , Rabdomiosarcoma Embrionario/genética , Sarcoglicanos/metabolismo , Análisis de Matrices Tisulares , Proteína p53 Supresora de Tumor/metabolismo
20.
Metab Brain Dis ; 26(1): 9-19, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21225451

RESUMEN

The primary pathology in mucopolysaccharidosis (MPS) IIIB is lysosomal storage of heparan sulfate (HS) glycosaminoglycans, leading to complex neuropathology and dysfunction, for which the detailed mechanisms remain unclear. Using antibodies that recognize specific HS glycoforms, we demonstrate differential cell-specific and domain-specific lysosomal HS-GAG distribution in MPS IIIB mouse brain. We also describe a novel neuron-specific brain HS epitope with broad, non-specific increase in the expression in all neurons in MPS IIIB mouse brain, including cerebellar granule neurons, which do not exhibit lysosomal storage pathology. This suggests that biosynthesis of certain HS glycoforms is enhanced throughout the CNS of MPS IIIB mice. Such a conclusion is further supported by demonstration of increased expression of multiple genes encoding enzymes essential in HS biosynthesis, including HS sulfotransferases and epimerases, as well as FGFs, for which HS serves as a co-receptor, in MPS IIIB brain. These data suggest that lysosomal storage of HS may lead to the increase in HS biosyntheses, which may contribute to the neuropathology of MPS IIIB by exacerbating the lysosomal HS storage.


Asunto(s)
Encéfalo , Heparitina Sulfato/biosíntesis , Lisosomas/metabolismo , Mucopolisacaridosis III , Animales , Encéfalo/enzimología , Encéfalo/patología , Carbohidrato Epimerasas/genética , Carbohidrato Epimerasas/metabolismo , Modelos Animales de Enfermedad , Factores de Crecimiento de Fibroblastos/genética , Factores de Crecimiento de Fibroblastos/metabolismo , Lisosomas/patología , Ratones , Ratones Noqueados , Mucopolisacaridosis III/enzimología , Mucopolisacaridosis III/genética , Mucopolisacaridosis III/patología , Neuronas/enzimología , Neuronas/patología , Isoformas de Proteínas/análisis , Sulfotransferasas/genética , Sulfotransferasas/metabolismo , Distribución Tisular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA