Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Nat Commun ; 13(1): 1847, 2022 04 14.
Artigo em Inglês | MEDLINE | ID: mdl-35422047

RESUMO

Ribitol-phosphate modification is crucial for the functional maturation of α-dystroglycan. Its dysfunction is associated with muscular dystrophy, cardiomyopathy, and central nervous system abnormalities; however, no effective treatments are currently available for diseases caused by ribitol-phosphate defects. In this study, we demonstrate that prodrug treatments can ameliorate muscular dystrophy caused by defects in isoprenoid synthase domain containing (ISPD), which encodes an enzyme that synthesizes CDP-ribitol, a donor substrate for ribitol-phosphate modification. We generated skeletal muscle-selective Ispd conditional knockout mice, leading to a pathogenic reduction in CDP-ribitol levels, abnormal glycosylation of α-dystroglycan, and severe muscular dystrophy. Adeno-associated virus-mediated gene replacement experiments suggested that the recovery of CDP-ribitol levels rescues the ISPD-deficient pathology. As a prodrug treatment strategy, we developed a series of membrane-permeable CDP-ribitol derivatives, among which tetraacetylated CDP-ribitol ameliorated the dystrophic pathology. In addition, the prodrug successfully rescued abnormal α-dystroglycan glycosylation in patient fibroblasts. Consequently, our findings provide proof-of-concept for supplementation therapy with CDP-ribitol and could accelerate the development of therapeutic agents for muscular dystrophy and other diseases caused by glycosylation defects.


Assuntos
Distrofias Musculares , Pró-Fármacos , Animais , Humanos , Camundongos , Modelos Animais de Doenças , Distroglicanas , Músculo Esquelético , Distrofias Musculares/tratamento farmacológico , Distrofias Musculares/genética , Fosfatos , Pró-Fármacos/farmacologia , Pró-Fármacos/uso terapêutico , Ribitol/uso terapêutico
2.
Biochem Biophys Res Commun ; 579: 8-14, 2021 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-34583196

RESUMO

α-Dystroglycan (α-DG) is a glycoprotein specifically modified with O-mannosyl glycans bearing long polysaccharides, termed matriglycans, which comprise repeating units of glucuronic acid and xylose. The matriglycan is linked to the O-mannosyl glycan core through two ribitol phosphate units that can be replaced with glycerol phosphate (GroP) units synthesized by fukutin and fukutin-related protein that transfer GroP from CDP-Gro. Here, we found that forced expression of the bacterial CDP-Gro synthase, TagD, from Bacillus subtilis could result in the overproduction of CDP-Gro in human colon carcinoma HCT116 cells. Western blot and liquid chromatography-tandem mass spectrometry analyses indicated that α-DG prepared from the TagD-expressing HCT116 cells contained abundant GroP and lacked matriglycans. Using the GroP-containing recombinant α-DG-Fc, we developed a novel monoclonal antibody, termed DG2, that reacts with several truncated glycoforms of α-DG, including GroP-terminated glycoforms lacking matriglycans; we verified the reactivity of DG2 against various types of knockout cells deficient in the biosynthesis of matriglycans. Accordingly, forced expression of TagD in HCT116 cells resulted in the reduction of matriglycans and an increase in DG2 reactivity. Collectively, our results indicate that DG2 could serve as a useful tool to determine tissue distribution and function of α-DG lacking matriglycans under physiological and pathophysiological conditions.


Assuntos
Anticorpos Monoclonais/química , Distroglicanas/química , Laminina/química , Isoformas de Proteínas/química , Animais , Bacillus subtilis , Sistemas CRISPR-Cas , Cromatografia Líquida , DNA Complementar/metabolismo , Feminino , Ácido Glucurônico/química , Glicopeptídeos/química , Células HCT116 , Humanos , Espectrometria de Massas , Camundongos , Camundongos Endogâmicos BALB C , Fosfatos , Polissacarídeos , Ligação Proteica , Conformação Proteica , Proteínas Recombinantes/química , Ribitol/química , Xilose
3.
EMBO Rep ; 20(11): e47967, 2019 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-31566294

RESUMO

Dystroglycan, an extracellular matrix receptor, has essential functions in various tissues. Loss of α-dystroglycan-laminin interaction due to defective glycosylation of α-dystroglycan underlies a group of congenital muscular dystrophies often associated with brain malformations, referred to as dystroglycanopathies. The lack of isogenic human dystroglycanopathy cell models has limited our ability to test potential drugs in a human- and neural-specific context. Here, we generated induced pluripotent stem cells (iPSCs) from a severe dystroglycanopathy patient with homozygous FKRP (fukutin-related protein gene) mutation. We showed that CRISPR/Cas9-mediated gene correction of FKRP restored glycosylation of α-dystroglycan in iPSC-derived cortical neurons, whereas targeted gene mutation of FKRP in wild-type cells disrupted this glycosylation. In parallel, we screened 31,954 small molecule compounds using a mouse myoblast line for increased glycosylation of α-dystroglycan. Using human FKRP-iPSC-derived neural cells for hit validation, we demonstrated that compound 4-(4-bromophenyl)-6-ethylsulfanyl-2-oxo-3,4-dihydro-1H-pyridine-5-carbonitrile (4BPPNit) significantly augmented glycosylation of α-dystroglycan, in part through upregulation of LARGE1 glycosyltransferase gene expression. Together, isogenic human iPSC-derived cells represent a valuable platform for facilitating dystroglycanopathy drug discovery and therapeutic development.


Assuntos
Avaliação Pré-Clínica de Medicamentos , Distroglicanas/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Sequência de Bases , Sistemas CRISPR-Cas , Células Cultivadas , Avaliação Pré-Clínica de Medicamentos/métodos , Distroglicanas/genética , Edição de Genes , Marcação de Genes , Loci Gênicos , Glicosilação/efeitos dos fármacos , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Imagem Molecular , Distrofias Musculares/tratamento farmacológico , Distrofias Musculares/etiologia , Distrofias Musculares/metabolismo , Mutação , N-Acetilglucosaminiltransferases/genética , N-Acetilglucosaminiltransferases/metabolismo , Células-Tronco Neurais/metabolismo , Neurônios/metabolismo , Pentosiltransferases/genética , Pentosiltransferases/metabolismo
4.
Clin Chem ; 65(10): 1295-1306, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31375477

RESUMO

BACKGROUND: Many muscular dystrophies currently remain untreatable. Recently, dietary ribitol has been suggested as a treatment for cytidine diphosphate (CDP)-l-ribitol pyrophosphorylase A (CRPPA, ISPD), fukutin (FKTN), and fukutin-related protein (FKRP) myopathy, by raising CDP-ribitol concentrations. Thus, to facilitate fast diagnosis, treatment development, and treatment monitoring, sensitive detection of CDP-ribitol is required. METHODS: An LC-MS method was optimized for CDP-ribitol in human and mice cells and tissues. RESULTS: CDP-ribitol, the product of CRPPA, was detected in all major human and mouse tissues. Moreover, CDP-ribitol concentrations were reduced in fibroblasts and skeletal muscle biopsies from patients with CRPPA myopathy, showing that CDP-ribitol could serve as a diagnostic marker to identify patients with CRPPA with severe Walker-Warburg syndrome and mild limb-girdle muscular dystrophy (LGMD) phenotypes. A screen for potentially therapeutic monosaccharides revealed that ribose, in addition to ribitol, restored CDP-ribitol concentrations and the associated O-glycosylation defect of α-dystroglycan. As the effect occurred in a mutation-dependent manner, we established a CDP-ribitol blood test to facilitate diagnosis and predict individualized treatment response. Ex vivo incubation of blood cells with ribose or ribitol restored CDP-ribitol concentrations in a patient with CRPPA LGMD. CONCLUSIONS: Sensitive detection of CDP-ribitol with LC-MS allows fast diagnosis of patients with severe and mild CRPPA myopathy. Ribose offers a readily testable dietary therapy for CRPPA myopathy, with possible applicability for patients with FKRP and FKTN myopathy. Evaluation of CDP-ribitol in blood is a promising tool for the evaluation and monitoring of dietary therapies for CRPPA myopathy in a patient-specific manner.


Assuntos
Monitoramento de Medicamentos/métodos , Distrofias Musculares/sangue , Distrofias Musculares/tratamento farmacológico , Açúcares de Nucleosídeo Difosfato/sangue , Animais , Cromatografia Líquida , Suplementos Nutricionais , Distroglicanas , Feminino , Glicosilação , Células HEK293 , Humanos , Masculino , Espectrometria de Massas , Camundongos , Camundongos Transgênicos , Pessoa de Meia-Idade , Músculo Esquelético/patologia , Distrofias Musculares/patologia , Mutação , Açúcares de Nucleosídeo Difosfato/análise , Nucleotidiltransferases/genética , Ribitol/farmacologia , Ribose/farmacologia
5.
J Mol Neurosci ; 58(2): 201-9, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26411569

RESUMO

Several dystrophin Dp71 messenger RNA (mRNA) alternative splice variants have been described. According to the splicing of exon 78 or intron 77, Dp71 proteins are grouped as Dp71d, Dp71f, and Dp71e, and each group has a specific C-terminal end. In this study, we explored the expression of Dp71 isoforms at the complementary DNA (cDNA) level and the subcellular localization of recombinant Myc-Dp71 proteins in PC12 cells. We determined that PC12 cells express Dp71a, Dp71c, Dp71ab, Dp71e, and Dp71ec mRNA splice variants. In undifferentiated and nerve growth factor-differentiated PC12 Tet-ON cells, Dp71a, Dp71ab, and Dp71e were found to localize and colocalize with ß-dystroglycan and α1-syntrophin in the periphery/cytoplasm, while Dp71c and Dp71ec were mainly localized in the cell periphery and showed less colocalization with ß-dystroglycan and α1-syntrophin. The levels of Dp71a, Dp71e, and Dp71ec were increased in the nucleus of differentiated PC12 Tet-ON cells compared to undifferentiated cells. Dp71 isoforms were also localized in neurite extensions and growth cones.


Assuntos
Proteínas de Ligação ao Cálcio/metabolismo , Distroglicanas/metabolismo , Proteínas de Membrana/metabolismo , Proteínas Musculares/metabolismo , Animais , Proteínas de Ligação ao Cálcio/genética , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Distroglicanas/genética , Distrofina/genética , Distrofina/metabolismo , Cones de Crescimento/metabolismo , Proteínas de Membrana/genética , Proteínas Musculares/genética , Células PC12 , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Transporte Proteico , Ratos
6.
Bioorg Med Chem ; 23(24): 7661-70, 2015 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-26652968

RESUMO

Alpha-dystroglycan (α-DG), a highly glycosylated receptor for extracellular matrix proteins, plays a critical role in many biological processes. Hypoglycosylation of α-DG results in various types of muscular dystrophies and is also highly associated with progression of majority of cancers. Currently, there are no effective treatments for those devastating diseases. Enhancing functional O-mannosyl glycans (FOG) of α-DG on the cell surfaces is a potential approach to address this unmet challenge. Based on the hypothesis that the cells can up-regulate FOG of α-DG in response to certain chemical stimuli, we developed a cell-based high-throughput screening (HTS) platform for searching chemical enhancers of FOG of α-DG from a large chemical library with 364,168 compounds. Sequential validation of the hits from a primary screening campaign and chemical works led to identification of a cluster of compounds that positively modulate FOG of α-DG on various cell surfaces including patient-derived myoblasts. These compounds enhance FOG of α-DG by almost ten folds, which provide us powerful tools for O-mannosylation studies and potential starting points for the development of drug to treat dystroglycanopathy.


Assuntos
Distroglicanas/metabolismo , Glicosilação/efeitos dos fármacos , Manose/metabolismo , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/farmacologia , Animais , Células CHO , Linhagem Celular , Cricetulus , Avaliação Pré-Clínica de Medicamentos/métodos , Ensaios de Triagem em Larga Escala/métodos , Humanos , Camundongos , Distrofias Musculares/tratamento farmacológico , Distrofias Musculares/metabolismo
7.
PLoS Biol ; 10(10): e1001409, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23109907

RESUMO

Muscular dystrophies are common, currently incurable diseases. A subset of dystrophies result from genetic disruptions in complexes that attach muscle fibers to their surrounding extracellular matrix microenvironment. Cell-matrix adhesions are exquisite sensors of physiological conditions and mediate responses that allow cells to adapt to changing conditions. Thus, one approach towards finding targets for future therapeutic applications is to identify cell adhesion pathways that mediate these dynamic, adaptive responses in vivo. We find that nicotinamide riboside kinase 2b-mediated NAD+ biosynthesis, which functions as a small molecule agonist of muscle fiber-extracellular matrix adhesion, corrects dystrophic phenotypes in zebrafish lacking either a primary component of the dystrophin-glycoprotein complex or integrin alpha7. Exogenous NAD+ or a vitamin precursor to NAD+ reduces muscle fiber degeneration and results in significantly faster escape responses in dystrophic embryos. Overexpression of paxillin, a cell adhesion protein downstream of NAD+ in this novel cell adhesion pathway, reduces muscle degeneration in zebrafish with intact integrin receptors but does not improve motility. Activation of this pathway significantly increases organization of laminin, a major component of the extracellular matrix basement membrane. Our results indicate that the primary protective effects of NAD+ result from changes to the basement membrane, as a wild-type basement membrane is sufficient to increase resilience of dystrophic muscle fibers to damage. The surprising result that NAD+ supplementation ameliorates dystrophy in dystrophin-glycoprotein complex- or integrin alpha7-deficient zebrafish suggests the existence of an additional laminin receptor complex that anchors muscle fibers to the basement membrane. We find that integrin alpha6 participates in this pathway, but either integrin alpha7 or the dystrophin-glycoprotein complex is required in conjunction with integrin alpha6 to reduce muscle degeneration. Taken together, these results define a novel cell adhesion pathway that may have future therapeutic relevance for a broad spectrum of muscular dystrophies.


Assuntos
Distrofias Musculares/metabolismo , NAD/biossíntese , Peixe-Zebra/metabolismo , Animais , Antígenos CD/genética , Antígenos CD/metabolismo , Adesão Celular , Modelos Animais de Doenças , Distroglicanas/genética , Distroglicanas/metabolismo , Distrofina/metabolismo , Matriz Extracelular/metabolismo , Cadeias alfa de Integrinas/genética , Cadeias alfa de Integrinas/metabolismo , Integrina alfa6/genética , Integrina alfa6/metabolismo , Laminina/metabolismo , Músculo Esquelético/metabolismo , Distrofias Musculares/genética , Paxilina/genética , Paxilina/metabolismo , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
8.
PLoS One ; 5(8): e12119, 2010 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-20711348

RESUMO

In the cerebellar cortex, interneurons of the molecular layer (stellate and basket cells) provide GABAergic input to Purkinje cells, as well as to each other and possibly to other interneurons. GABAergic inhibition in the molecular layer has mainly been investigated at the interneuron to Purkinje cell synapse. In this study, we used complementary subtractive strategies to quantitatively assess the ratio of GABAergic synapses on Purkinje cell dendrites versus those on interneurons. We generated a mouse model in which the GABAA receptor alpha1 subunit (GABAARalpha1) was selectively removed from Purkinje cells using the Cre/loxP system. Deletion of the alpha1 subunit resulted in a complete loss of GABAAR aggregates from Purkinje cells, allowing us to determine the density of GABAAR clusters in interneurons. In a complementary approach, we determined the density of GABA synapses impinging on Purkinje cells using alpha-dystroglycan as a specific marker of inhibitory postsynaptic sites. Combining these inverse approaches, we found that synapses received by interneurons represent approximately 40% of all GABAergic synapses in the molecular layer. Notably, this proportion was stable during postnatal development, indicating synchronized synaptogenesis. Based on the pure quantity of GABAergic synapses onto interneurons, we propose that mutual inhibition must play an important, yet largely neglected, computational role in the cerebellar cortex.


Assuntos
Córtex Cerebelar/citologia , Córtex Cerebelar/metabolismo , Sinapses/metabolismo , Ácido gama-Aminobutírico/metabolismo , Animais , Anticorpos/imunologia , Anticorpos/metabolismo , Distroglicanas/imunologia , Técnicas de Inativação de Genes , Camundongos , Neurônios/metabolismo , Subunidades Proteicas/deficiência , Subunidades Proteicas/genética , Células de Purkinje/metabolismo , Receptores de GABA-A/genética , Receptores de GABA-A/metabolismo , Deleção de Sequência , Especificidade por Substrato
9.
J Biol Chem ; 272(35): 22315-21, 1997 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-9268382

RESUMO

Because the polypeptide core of alpha-dystroglycan is encoded by a single gene, the difference in apparent molecular mass between alpha-dystroglycans expressed in various tissues is presumably due to differential glycosylation. However, little is presently known about the tissue-specific differences in alpha-dystroglycan glycosylation and whether these modifications may confer functional variability to alpha-dystroglycan. We recently observed that laminin-1 binding to skeletal muscle alpha-dystroglycan was dramatically inhibited by heparin, whereas the binding of commercial merosin to skeletal muscle alpha-dystroglycan was only marginally inhibited (Pall, E. A., Bolton, K. M., and Ervasti, J. M. (1996) J. Biol. Chem. 3817-3821). In contrast to 156-kDa skeletal muscle alpha-dystroglycan, both laminin-1 and merosin binding to 120-kDa brain alpha-dystroglycan were sensitive to heparin. We have now examined the laminin binding properties of 140-kDa alpha-dystroglycan purified from cardiac muscle and observed that like skeletal muscle alpha-dystroglycan, heparin inhibited cardiac alpha-dystroglycan binding to laminin-1, but not to merosin. On the other hand, cardiac and brain alpha-dystroglycans could be distinguished from skeletal muscle alpha-dystroglycan by their reactivity with the terminal GalNAc-specific lectin Vicia villosa agglutinin. Interestingly, skeletal muscle alpha-dystroglycan became reactive with V. villosa agglutinin upon digestion with sialidase from Clostridium perfringens, Arthrobacter neurofaciens, or Streptococcus, but not Vibrio cholerae or Newcastle disease virus sialidase. While none of the sialidase treatments affected the laminin binding properties of alpha-dystroglycan, the sum of our results suggests that skeletal muscle alpha-dystroglycan contains a novel sialic acid residue linked alpha2-6 to GalNAc. These properties are also consistent with the cellular characteristics of a GalNAc-terminated glycoconjugate recently implicated in neuromuscular synaptogenesis. Thus, variations in alpha-dystroglycan sialoglycosylation may prove as useful markers to further elucidate the role of alpha-dystroglycan glycoforms in different tissues and perhaps within a single cell type.


Assuntos
Proteínas do Citoesqueleto/metabolismo , Lectinas/metabolismo , Glicoproteínas de Membrana/metabolismo , Músculo Esquelético/metabolismo , Ácido N-Acetilneuramínico/metabolismo , Animais , Galinhas , Clostridium perfringens , Distroglicanas , Fabaceae , Heparina/farmacologia , Laminina/metabolismo , Miocárdio/metabolismo , Lectinas de Plantas , Plantas Medicinais , Coelhos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA