Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Am J Hum Genet ; 99(5): 1181-1189, 2016 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-27773428

RESUMO

Cobblestone lissencephaly (COB) is a severe brain malformation in which overmigration of neurons and glial cells into the arachnoid space results in the formation of cortical dysplasia. COB occurs in a wide range of genetic disorders known as dystroglycanopathies, which are congenital muscular dystrophies associated with brain and eye anomalies and range from Walker-Warburg syndrome to Fukuyama congenital muscular dystrophy. Each of these conditions has been associated with alpha-dystroglycan defects or with mutations in genes encoding basement membrane components, which are known to interact with alpha-dystroglycan. Our screening of a cohort of 25 families with recessive forms of COB identified six families affected by biallelic mutations in TMTC3 (encoding transmembrane and tetratricopeptide repeat containing 3), a gene without obvious functional connections to alpha-dystroglycan. Most affected individuals showed brainstem and cerebellum hypoplasia, as well as ventriculomegaly. However, the minority of the affected individuals had eye defects or elevated muscle creatine phosphokinase, separating the TMTC3 COB phenotype from typical congenital muscular dystrophies. Our data suggest that loss of TMTC3 causes COB with minimal eye or muscle involvement.


Assuntos
Alelos , Proteínas de Transporte/genética , Lissencefalia Cobblestone/genética , Proteínas de Membrana/genética , Sequência de Aminoácidos , Membrana Basal/metabolismo , Encéfalo/anormalidades , Encéfalo/diagnóstico por imagem , Proteínas de Transporte/metabolismo , Cerebelo/anormalidades , Cerebelo/diagnóstico por imagem , Lissencefalia Cobblestone/diagnóstico por imagem , Deficiências do Desenvolvimento/diagnóstico por imagem , Deficiências do Desenvolvimento/genética , Distroglicanas/metabolismo , Anormalidades do Olho/diagnóstico por imagem , Anormalidades do Olho/genética , Feminino , Humanos , Lactente , Masculino , Proteínas de Membrana/metabolismo , Mutação , Malformações do Sistema Nervoso/diagnóstico por imagem , Malformações do Sistema Nervoso/genética , Neuroglia/metabolismo , Neurônios/patologia , Linhagem , Fenótipo
2.
Proc Natl Acad Sci U S A ; 113(39): 10992-7, 2016 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-27625424

RESUMO

Dystroglycan (DG) is a highly expressed extracellular matrix receptor that is linked to the cytoskeleton in skeletal muscle. DG is critical for the function of skeletal muscle, and muscle with primary defects in the expression and/or function of DG throughout development has many pathological features and a severe muscular dystrophy phenotype. In addition, reduction in DG at the sarcolemma is a common feature in muscle biopsies from patients with various types of muscular dystrophy. However, the consequence of disrupting DG in mature muscle is not known. Here, we investigated muscles of transgenic mice several months after genetic knockdown of DG at maturity. In our study, an increase in susceptibility to contraction-induced injury was the first pathological feature observed after the levels of DG at the sarcolemma were reduced. The contraction-induced injury was not accompanied by increased necrosis, excitation-contraction uncoupling, or fragility of the sarcolemma. Rather, disruption of the sarcomeric cytoskeleton was evident as reduced passive tension and decreased titin immunostaining. These results reveal a role for DG in maintaining the stability of the sarcomeric cytoskeleton during contraction and provide mechanistic insight into the cause of the reduction in strength that occurs in muscular dystrophy after lengthening contractions.


Assuntos
Citoesqueleto/metabolismo , Distroglicanas/metabolismo , Contração Muscular , Músculo Esquelético/patologia , Músculo Esquelético/fisiopatologia , Sarcômeros/metabolismo , Animais , Conectina/metabolismo , Citoesqueleto/efeitos dos fármacos , Acoplamento Excitação-Contração/efeitos dos fármacos , Feminino , Contração Isométrica/efeitos dos fármacos , Masculino , Camundongos Knockout , Contração Muscular/efeitos dos fármacos , Músculo Esquelético/efeitos dos fármacos , Necrose , Tamanho do Órgão , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Sarcolema/metabolismo , Sarcômeros/efeitos dos fármacos , Tamoxifeno/farmacologia
3.
Am J Hum Genet ; 92(3): 354-65, 2013 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-23453667

RESUMO

Mutations in several known or putative glycosyltransferases cause glycosylation defects in α-dystroglycan (α-DG), an integral component of the dystrophin glycoprotein complex. The hypoglycosylation reduces the ability of α-DG to bind laminin and other extracellular matrix ligands and is responsible for the pathogenesis of an inherited subset of muscular dystrophies known as the dystroglycanopathies. By exome and Sanger sequencing we identified two individuals affected by a dystroglycanopathy with mutations in ß-1,3-N-acetylgalactosaminyltransferase 2 (B3GALNT2). B3GALNT2 transfers N-acetyl galactosamine (GalNAc) in a ß-1,3 linkage to N-acetyl glucosamine (GlcNAc). A subsequent study of a separate cohort of individuals identified recessive mutations in four additional cases that were all affected by dystroglycanopathy with structural brain involvement. We show that functional dystroglycan glycosylation was reduced in the fibroblasts and muscle (when available) of these individuals via flow cytometry, immunoblotting, and immunocytochemistry. B3GALNT2 localized to the endoplasmic reticulum, and this localization was perturbed by some of the missense mutations identified. Moreover, knockdown of b3galnt2 in zebrafish recapitulated the human congenital muscular dystrophy phenotype with reduced motility, brain abnormalities, and disordered muscle fibers with evidence of damage to both the myosepta and the sarcolemma. Functional dystroglycan glycosylation was also reduced in the b3galnt2 knockdown zebrafish embryos. Together these results demonstrate a role for B3GALNT2 in the glycosylation of α-DG and show that B3GALNT2 mutations can cause dystroglycanopathy with muscle and brain involvement.


Assuntos
Distroglicanas/genética , Distrofias Musculares/genética , Mutação , N-Acetilgalactosaminiltransferases/genética , Animais , Encéfalo/enzimologia , Encéfalo/metabolismo , Linhagem Celular , Distroglicanas/metabolismo , Retículo Endoplasmático/enzimologia , Retículo Endoplasmático/genética , Retículo Endoplasmático/metabolismo , Feminino , Fibroblastos/enzimologia , Fibroblastos/metabolismo , Predisposição Genética para Doença , Glicosilação , Humanos , Lactente , Masculino , Músculo Esquelético/enzimologia , Músculo Esquelético/metabolismo , Distrofias Musculares/enzimologia , Distrofias Musculares/metabolismo , N-Acetilgalactosaminiltransferases/metabolismo , Peixe-Zebra
4.
Am J Hum Genet ; 93(1): 29-41, 2013 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-23768512

RESUMO

Congenital muscular dystrophies with hypoglycosylation of α-dystroglycan (α-DG) are a heterogeneous group of disorders often associated with brain and eye defects in addition to muscular dystrophy. Causative variants in 14 genes thought to be involved in the glycosylation of α-DG have been identified thus far. Allelic mutations in these genes might also cause milder limb-girdle muscular dystrophy phenotypes. Using a combination of exome and Sanger sequencing in eight unrelated individuals, we present evidence that mutations in guanosine diphosphate mannose (GDP-mannose) pyrophosphorylase B (GMPPB) can result in muscular dystrophy variants with hypoglycosylated α-DG. GMPPB catalyzes the formation of GDP-mannose from GTP and mannose-1-phosphate. GDP-mannose is required for O-mannosylation of proteins, including α-DG, and it is the substrate of cytosolic mannosyltransferases. We found reduced α-DG glycosylation in the muscle biopsies of affected individuals and in available fibroblasts. Overexpression of wild-type GMPPB in fibroblasts from an affected individual partially restored glycosylation of α-DG. Whereas wild-type GMPPB localized to the cytoplasm, five of the identified missense mutations caused formation of aggregates in the cytoplasm or near membrane protrusions. Additionally, knockdown of the GMPPB ortholog in zebrafish caused structural muscle defects with decreased motility, eye abnormalities, and reduced glycosylation of α-DG. Together, these data indicate that GMPPB mutations are responsible for congenital and limb-girdle muscular dystrophies with hypoglycosylation of α-DG.


Assuntos
Distroglicanas/metabolismo , Distrofia Muscular do Cíngulo dos Membros/genética , Mutação de Sentido Incorreto , Nucleotidiltransferases/metabolismo , Animais , Pré-Escolar , Análise Mutacional de DNA/métodos , Distroglicanas/genética , Anormalidades do Olho/patologia , Feminino , Fibroblastos/enzimologia , Fibroblastos/patologia , Estudos de Associação Genética/métodos , Glicosilação , Guanosina Difosfato Manose/metabolismo , Heterozigoto , Humanos , Lactente , Recém-Nascido , Masculino , Músculo Esquelético/enzimologia , Músculo Esquelético/patologia , Distrofia Muscular do Cíngulo dos Membros/enzimologia , Nucleotidiltransferases/genética , Peixe-Zebra/genética , Peixe-Zebra/metabolismo
5.
Proc Natl Acad Sci U S A ; 110(52): 21024-9, 2013 Dec 24.
Artigo em Inglês | MEDLINE | ID: mdl-24297939

RESUMO

In recent years protein O-mannosylation has become a focus of attention as a pathomechanism underlying severe congenital muscular dystrophies associated with neuronal migration defects. A key feature of these disorders is the lack of O-mannosyl glycans on α-dystroglycan, resulting in abnormal basement membrane formation. Additional functions of O-mannosylation are still largely unknown. Here, we identify the essential cell-cell adhesion glycoprotein epithelial (E)-cadherin as an O-mannosylated protein and establish a functional link between O-mannosyl glycans and cadherin-mediated cell-cell adhesion. By genetically and pharmacologically blocking protein O-mannosyltransferases, we found that this posttranslational modification is essential for preimplantation development of the mouse embryo. O-mannosylation-deficient embryos failed to proceed from the morula to the blastocyst stage because of defects in the molecular architecture of cell-cell contact sites, including the adherens and tight junctions. Using mass spectrometry, we demonstrate that O-mannosyl glycans are present on E-cadherin, the major cell-adhesion molecule of blastomeres, and present evidence that this modification is generally conserved in cadherins. Further, the use of newly raised antibodies specific for an O-mannosyl-conjugated epitope revealed that these glycans are present on early mouse embryos. Finally, our cell-aggregation assays demonstrated that O-mannosyl glycans are crucial for cadherin-based cell adhesion. Our results redefine the significance of O-mannosylation in humans and other mammals, showing the immense impact of cadherins on normal as well as pathogenic cell behavior.


Assuntos
Junções Aderentes/metabolismo , Caderinas/metabolismo , Adesão Celular/fisiologia , Embrião de Mamíferos/citologia , Desenvolvimento Embrionário/fisiologia , Manose/metabolismo , Animais , Primers do DNA/genética , Cães , Embrião de Mamíferos/fisiologia , Imunofluorescência , Glicosilação , Células Madin Darby de Rim Canino , Espectrometria de Massas , Camundongos , Polissacarídeos/metabolismo
6.
Hum Mutat ; 36(12): 1159-63, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26310427

RESUMO

Mutations in GDP-mannose pyrophosphorylase B (GMPPB), a catalyst for the formation of the sugar donor GDP-mannose, were recently identified as a cause of muscular dystrophy resulting from abnormal glycosylation of α-dystroglycan. In this series, we report nine unrelated individuals with GMPPB-associated dystroglycanopathy. The most mildly affected subject has normal strength at 25 years, whereas three severely affected children presented in infancy with intellectual disability and epilepsy. Muscle biopsies of all subjects are dystrophic with abnormal immunostaining for glycosylated α-dystroglycan. This cohort, together with previously published cases, allows preliminary genotype-phenotype correlations to be made for the emerging GMPPB common variants c.79G>C (p.D27H) and c.860G>A (p.R287Q). We observe that c.79G>C (p.D27H) is associated with a mild limb-girdle muscular dystrophy phenotype, whereas c.860G>A (p.R287Q) is associated with a relatively severe congenital muscular dystrophy typically involving brain development. Sixty-six percent of GMPPB families to date have one of these common variants.


Assuntos
Distroglicanas/metabolismo , Distrofias Musculares/genética , Distrofias Musculares/metabolismo , Mutação , Nucleotidiltransferases/genética , Fenótipo , Adolescente , Alelos , Biópsia , Encéfalo/patologia , Criança , Pré-Escolar , Feminino , Estudos de Associação Genética , Heterozigoto , Humanos , Lactente , Imageamento por Ressonância Magnética , Masculino , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Distrofias Musculares/diagnóstico , Adulto Jovem
7.
J Biol Chem ; 289(41): 28138-48, 2014 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-25138275

RESUMO

Mutations in the LARGE gene have been identified in congenital muscular dystrophy (CMD) patients with brain abnormalities. Both LARGE and its paralog, LARGE2 (also referred to as GYLTL1B) are bifunctional glycosyltransferases with xylosyltransferase (Xyl-T) and glucuronyltransferase (GlcA-T) activities, and are capable of forming polymers consisting of [-3Xyl-α1,3GlcAß1-] repeats. LARGE-dependent modification of α-dystroglycan (α-DG) with these polysaccharides is essential for the ability of α-DG to act as a receptor for ligands in the extracellular matrix. Here we report on the endogenous enzymatic activities of LARGE and LARGE2 in mice and humans, using a newly developed assay for GlcA-T activity. We show that normal mouse and human cultured cells have endogenous LARGE GlcA-T, and that this activity is absent in cells from the Large(myd) (Large-deficient) mouse model of muscular dystrophy, as well as in cells from CMD patients with mutations in the LARGE gene. We also demonstrate that GlcA-T activity is significant in the brain, heart, and skeletal muscle of wild-type and Large2(-/-) mice, but negligible in the corresponding tissues of the Large(myd) mice. Notably, GlcA-T activity is substantial, though reduced, in the kidneys of both the Large(myd) and Large2(-/-) mice, consistent with the observation of α-DG/laminin binding in these contexts. This study is the first to test LARGE activity in samples as small as cryosections and, moreover, provides the first direct evidence that not only LARGE, but also LARGE2, is vital to effective functional modification of α-DG in vivo.


Assuntos
Distroglicanas/metabolismo , Glicosiltransferases/metabolismo , Laminina/metabolismo , Distrofias Musculares/enzimologia , N-Acetilglucosaminiltransferases/metabolismo , Animais , Sítios de Ligação , Encéfalo/enzimologia , Encéfalo/patologia , Células Cultivadas , Criança , Modelos Animais de Doenças , Distroglicanas/genética , Ensaios Enzimáticos , Feminino , Fibroblastos/enzimologia , Fibroblastos/patologia , Regulação da Expressão Gênica , Glicosiltransferases/genética , Humanos , Rim/enzimologia , Rim/patologia , Laminina/genética , Camundongos , Camundongos Knockout , Músculo Esquelético/enzimologia , Músculo Esquelético/patologia , Distrofias Musculares/genética , Distrofias Musculares/patologia , Miocárdio/enzimologia , Miocárdio/patologia , N-Acetilglucosaminiltransferases/genética , Especificidade de Órgãos , Ligação Proteica
8.
N Engl J Med ; 364(10): 939-46, 2011 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-21388311

RESUMO

Dystroglycan, which serves as a major extracellular matrix receptor in muscle and the central nervous system, requires extensive O-glycosylation to function. We identified a dystroglycan missense mutation (Thr192→Met) in a woman with limb-girdle muscular dystrophy and cognitive impairment. A mouse model harboring this mutation recapitulates the immunohistochemical and neuromuscular abnormalities observed in the patient. In vitro and in vivo studies showed that the mutation impairs the receptor function of dystroglycan in skeletal muscle and brain by inhibiting the post-translational modification, mediated by the glycosyltransferase LARGE, of the phosphorylated O-mannosyl glycans on α-dystroglycan that is required for high-affinity binding to laminin.


Assuntos
Distroglicanas/genética , Distrofia Muscular do Cíngulo dos Membros/genética , Mutação de Sentido Incorreto , Animais , Modelos Animais de Doenças , Feminino , Humanos , Camundongos , Linhagem , Fenótipo , Análise de Sequência de DNA
9.
Brain ; 136(Pt 1): 269-81, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23288328

RESUMO

Dystroglycanopathies are a clinically and genetically diverse group of recessively inherited conditions ranging from the most severe of the congenital muscular dystrophies, Walker-Warburg syndrome, to mild forms of adult-onset limb-girdle muscular dystrophy. Their hallmark is a reduction in the functional glycosylation of α-dystroglycan, which can be detected in muscle biopsies. An important part of this glycosylation is a unique O-mannosylation, essential for the interaction of α-dystroglycan with extracellular matrix proteins such as laminin-α2. Mutations in eight genes coding for proteins in the glycosylation pathway are responsible for ∼50% of dystroglycanopathy cases. Despite multiple efforts using traditional positional cloning, the causative genes for unsolved dystroglycanopathy cases have escaped discovery for several years. In a recent collaborative study, we discovered that loss-of-function recessive mutations in a novel gene, called isoprenoid synthase domain containing (ISPD), are a relatively common cause of Walker-Warburg syndrome. In this article, we report the involvement of the ISPD gene in milder dystroglycanopathy phenotypes ranging from congenital muscular dystrophy to limb-girdle muscular dystrophy and identified allelic ISPD variants in nine cases belonging to seven families. In two ambulant cases, there was evidence of structural brain involvement, whereas in seven, the clinical manifestation was restricted to a dystrophic skeletal muscle phenotype. Although the function of ISPD in mammals is not yet known, mutations in this gene clearly lead to a reduction in the functional glycosylation of α-dystroglycan, which not only causes the severe Walker-Warburg syndrome but is also a common cause of the milder forms of dystroglycanopathy.


Assuntos
Distrofias Musculares/congênito , Distrofias Musculares/genética , Mutação , Nucleotidiltransferases/genética , Adolescente , Criança , Pré-Escolar , Distroglicanas/genética , Distroglicanas/metabolismo , Feminino , Glicosilação , Humanos , Imageamento por Ressonância Magnética , Masculino , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Distrofias Musculares/metabolismo , Distrofias Musculares/patologia , Distrofia Muscular do Cíngulo dos Membros/genética , Distrofia Muscular do Cíngulo dos Membros/metabolismo , Distrofia Muscular do Cíngulo dos Membros/patologia , Adulto Jovem
10.
Glycobiology ; 23(3): 295-302, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23125099

RESUMO

LARGE-dependent modification enables α-dystroglycan (α-DG) to bind to its extracellular matrix ligands. Mutations in the LARGE gene and several others involved in O-mannosyl glycan synthesis have been identified in congenital and limb-girdle muscular dystrophies that are characterized by perturbed glycosylation and reduced ligand-binding affinity of α-DG. LARGE is a bifunctional glycosyltransferase that alternately transfers xylose and glucuronic acid, thereby generating the heteropolysaccharides on α-DG that confer its ligand binding. Although the LARGE paralog LARGE2 (also referred to as GYLTL1B) has likewise been shown to enhance the functional modification of α-DG in cultured cells, its enzymatic activities have not been identified. Here, we report that LARGE2 is also a bifunctional glycosyltransferase and compare its properties with those of LARGE. By means of a high-performance liquid chromatography-based enzymatic assay, we demonstrate that like LARGE, LARGE2 has xylosyltransferase (Xyl-T) and glucuronyltransferase (GlcA-T) activities, as well as polymerizing activity. Notably, however, the pH optima of the Xyl-T and GlcA-T of LARGE2 are distinct from one another and also from those of LARGE. Our results suggest that LARGE and LARGE2 catalyze the same glycosylation reactions for the functional modification of α-DG, but that they have different biochemical properties.


Assuntos
Distroglicanas/metabolismo , Glicosiltransferases/metabolismo , N-Acetilglucosaminiltransferases/metabolismo , Animais , Células CHO , Domínio Catalítico , Cricetinae , Cricetulus , Ácido Glucurônico/metabolismo , Glicosiltransferases/química , Concentração de Íons de Hidrogênio , Cinética , Camundongos , N-Acetilglucosaminiltransferases/química , Multimerização Proteica , Xilose/metabolismo
11.
Mol Genet Metab ; 110(3): 345-351, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23856421

RESUMO

Congenital disorders of glycosylation (CDG) are rare genetic defects mainly in the post-translational modification of proteins via attachment of carbohydrate chains. We describe an infant with the phenotype of a congenital muscular dystrophy, with borderline microcephaly, hypotonia, camptodactyly, severe motor delay, and elevated creatine kinase. Muscle biopsy showed muscular dystrophy and reduced α-dystroglycan immunostaining with glycoepitope-specific antibodies in a pattern diagnostic of dystroglycanopathy. Carbohydrate deficient transferrin testing showed a pattern pointing to a CDG type I. Sanger sequencing of DPM1 (dolichol-P-mannose synthase subunit 1) revealed a novel Gly > Val change c.455G > T missense mutation resulting in p.Gly152Val) of unknown pathogenicity and deletion/duplication analysis revealed an intragenic deletion from exons 3 to 7 on the other allele. DPM1 activity in fibroblasts was reduced by 80%, while affinity for the substrate was not depressed, suggesting a decrease in the amount of active enzyme. Transfected cells expressing tagged versions of wild type and the p.Gly152Val mutant displayed reduced binding to DPM3, an essential, non-catalytic subunit of the DPM complex, suggesting a mechanism for pathogenicity. The present case is the first individual described with DPM1-CDG (CDG-Ie) to also have clinical and muscle biopsy findings consistent with dystroglycanopathy.


Assuntos
Defeitos Congênitos da Glicosilação/diagnóstico , Defeitos Congênitos da Glicosilação/genética , Manosiltransferases/genética , Distrofias Musculares/diagnóstico , Mutação , Biópsia , Diagnóstico Diferencial , Progressão da Doença , Ativação Enzimática , Éxons , Feminino , Ordem dos Genes , Humanos , Lactente , Masculino , Manosiltransferases/metabolismo , Músculo Esquelético/patologia
12.
Biomolecules ; 13(8)2023 08 07.
Artigo em Inglês | MEDLINE | ID: mdl-37627292

RESUMO

The treatment landscape for lysosomal storage disorders (LSDs) is rapidly evolving. An increase in the number of preclinical and clinical studies in the last decade has demonstrated that pharmacological chaperones are a feasible alternative to enzyme replacement therapy (ERT) for individuals with LSDs. A systematic search was performed to retrieve and critically assess the evidence from preclinical and clinical applications of pharmacological chaperones in the treatment of LSDs and to elucidate the mechanisms by which they could be effective in clinical practice. Publications were screened according to the Preferred Reporting Items for Systematic reviews and Meta-Analyses (PRISMA) reporting guidelines. Fifty-two articles evaluating 12 small molecules for the treatment of seven LSDs are included in this review. Overall, a substantial amount of preclinical and clinical data support the potential of pharmacological chaperones as treatments for Fabry disease, Gaucher disease, and Pompe disease. Most of the available clinical evidence evaluated migalastat for the treatment of Fabry disease. There was a lack of consistency in the terminology used to describe pharmacological chaperones in the literature. Therefore, the new small molecule chaperone (SMC) classification system is proposed to inform a standardized approach for new, emerging small molecule therapies in LSDs.


Assuntos
Doença de Fabry , Doença de Gaucher , Doenças por Armazenamento dos Lisossomos , Humanos , Doenças por Armazenamento dos Lisossomos/tratamento farmacológico , Doença de Fabry/tratamento farmacológico , Doença de Gaucher/tratamento farmacológico , Terapia de Reposição de Enzimas , Lisossomos
13.
Nat Commun ; 13(1): 3617, 2022 06 24.
Artigo em Inglês | MEDLINE | ID: mdl-35750689

RESUMO

α-Dystroglycan (α-DG) is uniquely modified on O-mannose sites by a repeating disaccharide (-Xylα1,3-GlcAß1,3-)n termed matriglycan, which is a receptor for laminin-G domain-containing proteins and employed by old-world arenaviruses for infection. Using chemoenzymatically synthesized matriglycans printed as a microarray, we demonstrate length-dependent binding to Laminin, Lassa virus GP1, and the clinically-important antibody IIH6. Utilizing an enzymatic engineering approach, an N-linked glycoprotein was converted into a IIH6-positive Laminin-binding glycoprotein. Engineering of the surface of cells deficient for either α-DG or O-mannosylation with matriglycans of sufficient length recovers infection with a Lassa-pseudovirus. Finally, free matriglycan in a dose and length dependent manner inhibits viral infection of wildtype cells. These results indicate that matriglycan alone is necessary and sufficient for IIH6 staining, Laminin and LASV GP1 binding, and Lassa-pseudovirus infection and support a model in which it is a tunable receptor for which increasing chain length enhances ligand-binding capacity.


Assuntos
Distroglicanas , Laminina , Distroglicanas/metabolismo , Glicoproteínas/metabolismo , Laminina/metabolismo , Vírus Lassa/metabolismo , Polissacarídeos/metabolismo
14.
J Neurosci ; 28(42): 10567-75, 2008 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-18923033

RESUMO

Walker-Warburg syndrome (WWS) is a severe congenital disease that is characterized by brain and eye malformations and lethality during the first year of life. Genetic mutations have been identified in a subset of WWS patients, but a majority of clinical cases have unknown etiologies. POMT1 and POMT2, two of the causative genes, form an active enzyme complex in the posttranslational biosynthetic pathway of dystroglycan. Deletion of either Pomt1 or the dystroglycan gene causes early embryonic lethality in mice. Here we report that mice with epiblast-specific loss of dystroglycan develop brain and eye defects that broadly resemble the clinical spectrum of the human disease, including aberrant neuron migration, hydrocephalus, and malformations of the anterior and posterior chambers of the eye. Breaches of basement membranes coincide with the pathology, revealing an important function for dystroglycan in the morphogenesis of the brain and eye. These findings demonstrate the central role of dystroglycan in WWS and suggest that novel defects in posttranslational processing or mutations of the dystroglycan gene itself may underlie cases in which no causative mutation has been found.


Assuntos
Encéfalo/anormalidades , Encéfalo/metabolismo , Distroglicanas/deficiência , Anormalidades do Olho/metabolismo , Camadas Germinativas/anormalidades , Camadas Germinativas/metabolismo , Anormalidades Múltiplas/genética , Anormalidades Múltiplas/metabolismo , Anormalidades Múltiplas/patologia , Animais , Encéfalo/patologia , Distroglicanas/genética , Anormalidades do Olho/genética , Anormalidades do Olho/patologia , Camadas Germinativas/patologia , Humanos , Hidrocefalia/genética , Hidrocefalia/metabolismo , Hidrocefalia/patologia , Lactente , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Síndrome
15.
Glycobiology ; 18(8): 615-25, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18490429

RESUMO

O-Mannosylation represents an evolutionarily conserved, essential protein modification. In mammals the protein O-mannosyltransferases POMT1 and POMT2 act as a heteromeric complex to initiate O-mannosylation in the endoplasmic reticulum. Mutations in human POMT1 and POMT2 cause a group of congenital muscular dystrophies due to reduced O-glycosylation of alpha-dystroglycan. The most severe of these autosomal recessive conditions is Walker-Warburg syndrome (WWS) with severe brain and ocular involvement. We previously showed in the murine model that Pomt1 is expressed in WWS-related tissues both during embryogenesis and in adults. Whereas there is only a single Pomt1 transcript in adult mice, we demonstrated that there are two Pomt2 transcripts, somatic sPomt2 and testis-specific tPomt2. In this study we demonstrate that sPomt2, but not tPomt2, is prominently expressed in mouse embryos in the tissues that are most severely affected in WWS (developing muscle, eye, and brain). Correlation of POMT transcripts and protein isoforms with POMT mannosyltransferase enzyme activity demonstrates that sPOMT2-POMT1 complexes catalyze mannosyltransfer in adult somatic tissues and testis. It is suggested that the gonadal defects described in some WWS cases are associated with defects in O-mannosylation. Our data further show that whereas sPOMT2 is widely expressed, tPOMT2 is restricted to the acrosome of male germ cells and is not involved in the biosynthesis of O-mannosyl glycans in vivo. We prove that tPOMT2 is highly conserved among mammals, including humans, suggesting a crucial function that is distinct from sPOMT2.


Assuntos
Encéfalo/anormalidades , Encéfalo/enzimologia , Manosiltransferases/metabolismo , Acrossomo/enzimologia , Envelhecimento/fisiologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Linhagem Celular , Sequência Conservada , Embrião de Mamíferos/embriologia , Embrião de Mamíferos/enzimologia , Regulação da Expressão Gênica no Desenvolvimento , Regulação Enzimológica da Expressão Gênica , Humanos , Isoenzimas/metabolismo , Masculino , Manosiltransferases/genética , Camundongos , Dados de Sequência Molecular , Especificidade de Órgãos , RNA Mensageiro/genética , Alinhamento de Sequência , Espermatogênese , Espermatozoides/citologia , Espermatozoides/enzimologia , Síndrome , Testículo/enzimologia
16.
Neuromuscul Disord ; 28(7): 592-596, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29759639

RESUMO

Mutations in POMT2 are most commonly associated with Walker-Warburg syndrome and Muscle-Eye-Brain disease, but can also cause limb girdle muscular dystrophy (LGMD2N). We report a case of LGMD due to a novel mutation in POMT2 unmasked by uniparental isodisomy. The patient experienced proximal muscle weakness from three years of age with minimal progression. She developed progressive contractures and underwent unilateral Achilles tenotomy. By age 11, she had borderline low left ventricular ejection fraction and mild restrictive lung disease. Muscle biopsy showed mild dystrophic changes with selective reduction in α-dystroglycan immunostaining. Sequencing of POMT2 showed a novel homozygous c.1502A>C variant that was predicted to be probably pathogenic. Fibroblast complementation studies showed lack of functional glycosylation rescued by wild-type POMT2 expression. Chromosomal microarray showed a single 15 Mb copy number neutral loss of heterozygosity on chromosome 14 encompassing POMT2. RNAseq verified homozygosity at this locus. Together, our findings indicate maternal uniparental isodisomy causing LGMD2N.


Assuntos
Manosiltransferases/genética , Distrofia Muscular do Cíngulo dos Membros/genética , Mutação , Dissomia Uniparental , Adolescente , Distroglicanas/metabolismo , Feminino , Humanos
17.
Curr Opin Struct Biol ; 13(5): 621-30, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-14568618

RESUMO

In yeasts and other fungi, O-mannosyl glycans constitute a major protein modification that is essential for cell viability. For several decades, protein O-mannosylation was considered a yeast-specific modification. Thus, it was especially interesting when it became evident that O-mannosyl glycans in mammals are not as rare as previously thought. O-mannosyl glycans are abundant in the mammalian brain and are also an abundant modification of alpha-dystroglycan, a component of the dystrophin-glycoprotein complex. Recently, mutations in genes that are or might be involved in the glycosylation of alpha-dystroglycan have been identified. Their association with neuromuscular diseases has focused the attention of different research areas on protein O-mannosylation.


Assuntos
Glicoproteínas/química , Glicoproteínas/metabolismo , Distrofias Musculares/metabolismo , Polissacarídeos/química , Polissacarídeos/metabolismo , Leveduras/química , Leveduras/metabolismo , Animais , Humanos , Mamíferos , Manose/química , Manose/metabolismo , Manosiltransferases/química , Manosiltransferases/metabolismo , Conformação Molecular , Estrutura Molecular , Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/metabolismo , Especificidade da Espécie , Relação Estrutura-Atividade
18.
Nat Commun ; 8: 15153, 2017 04 28.
Artigo em Inglês | MEDLINE | ID: mdl-28452368

RESUMO

Growth and differentiation factor 8 (GDF8) is a TGF-ß superfamily member, and negative regulator of skeletal muscle mass. GDF8 inhibition results in prominent muscle growth in mice, but less impressive hypertrophy in primates, including man. Broad TGF-ß inhibition suggests another family member negatively regulates muscle mass, and its blockade enhances muscle growth seen with GDF8-specific inhibition. Here we show that activin A is the long-sought second negative muscle regulator. Activin A specific inhibition, on top of GDF8 inhibition, leads to pronounced muscle hypertrophy and force production in mice and monkeys. Inhibition of these two ligands mimics the hypertrophy seen with broad TGF-ß blockers, while avoiding the adverse effects due to inhibition of multiple family members. Altogether, we identify activin A as a second negative regulator of muscle mass, and suggest that inhibition of both ligands provides a preferred therapeutic approach, which maximizes the benefit:risk ratio for muscle diseases in man.


Assuntos
Ativinas/metabolismo , Hipertrofia/patologia , Hipotonia Muscular/patologia , Músculo Esquelético/crescimento & desenvolvimento , Miostatina/metabolismo , Receptores de Activinas Tipo II/metabolismo , Ativinas/antagonistas & inibidores , Animais , Anticorpos Monoclonais/farmacologia , Índice de Massa Corporal , Dexametasona/farmacologia , Humanos , Contração Isométrica/fisiologia , Macaca fascicularis , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos SCID , Músculo Esquelético/fisiologia , Miostatina/antagonistas & inibidores , Ratos
19.
Elife ; 52016 04 29.
Artigo em Inglês | MEDLINE | ID: mdl-27130732

RESUMO

Multiple glycosyltransferases are essential for the proper modification of alpha-dystroglycan, as mutations in the encoding genes cause congenital/limb-girdle muscular dystrophies. Here we elucidate further the structure of an O-mannose-initiated glycan on alpha-dystroglycan that is required to generate its extracellular matrix-binding polysaccharide. This functional glycan contains a novel ribitol structure that links a phosphotrisaccharide to xylose. ISPD is a CDP-ribitol (ribose) pyrophosphorylase that generates the reduced sugar nucleotide for the insertion of ribitol in a phosphodiester linkage to the glycoprotein. TMEM5 is a UDP-xylosyl transferase that elaborates the structure. We demonstrate in a zebrafish model as well as in a human patient that defects in TMEM5 result in muscular dystrophy in combination with abnormal brain development. Thus, we propose a novel structure-a ribitol in a phosphodiester linkage-for the moiety on which TMEM5, B4GAT1, and LARGE act to generate the functional receptor for ECM proteins having LG domains.


Assuntos
Distroglicanas/química , Distroglicanas/metabolismo , Matriz Extracelular/metabolismo , Proteínas de Membrana/metabolismo , Polissacarídeos/análise , Animais , Humanos , Manose/análise , Nucleotidiltransferases/metabolismo , Pentosiltransferases , Ligação Proteica , Ribitol/análise , Peixe-Zebra
20.
Neuromuscul Disord ; 15(4): 271-5, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15792865

RESUMO

Mutations of the protein O-mannosyltransferase (POMT1) gene affect glycosylation of alpha-dystroglycan, leading to Walker-Warburg syndrome, a lethal disorder in early life with severe congenital muscular dystrophy, and brain and eye malformations. Recently, we described a novel form of recessive limb girdle muscular dystrophy with mild mental retardation, associated with an abnormal alpha-dystroglycan pattern in the muscle, suggesting a glycosylation defect. Here, we present evidence that this distinct phenotype results from a common mutation (A200P) in the POMT1 gene. Our findings further expand the phenotype of glycosylation disorders linked to POMT1 mutations. Furthermore, the A200P mutation is part of a conserved core haplotype, indicating an ancestral founder mutation.


Assuntos
Deficiência Intelectual/genética , Manosiltransferases/genética , Distrofia Muscular do Cíngulo dos Membros/genética , Mutação/genética , Adolescente , Adulto , Alanina/genética , Alelos , Criança , Análise Mutacional de DNA/métodos , Feminino , Humanos , Deficiência Intelectual/fisiopatologia , Masculino , Modelos Moleculares , Distrofia Muscular do Cíngulo dos Membros/fisiopatologia , Fenótipo , Polimorfismo de Nucleotídeo Único , Prolina/genética , RNA Mensageiro/biossíntese , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA