Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Hum Mol Genet ; 26(16): 3081-3093, 2017 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-28505249

RESUMO

Sialic acids are monosaccharides found in terminal sugar chains of cell surfaces and proteins; they have various biological functions and have been implicated in health and disease. Genetic defects of the GNE gene which encodes a critical bifunctional enzyme for sialic acid biosynthesis, lead to GNE myopathy, a disease manifesting with progressive muscle atrophy and weakness. The likely mechanism of disease is a lack of sialic acids. There remains, however, an unexplained link between hyposialylation and the muscle atrophy and weakness. In this study, we found that muscle proteins were highly modified by S-nitrosylation, and that oxidative stress-responsive genes were significantly upregulated, in hyposialylated muscles from human GNE myopathy patients and model mice. In both in vitro and in vivo models, the production of reactive oxygen species (ROS) was elevated with cellular hyposialylation, and increasing overall sialylation by extrinsic sialic acid intake reduced ROS and protein S-nitrosylation. More importantly, the antioxidant, oral N-acetylcysteine led to amelioration of the muscle atrophy and weakness in Gne mutant mice. Our data provide evidence of additional important function of sialic acids as a ROS scavenger in skeletal muscles, expanding our understanding on how sialic acid deficiency contributes to disease pathology, and identify oxidative stress as a therapeutic target in GNE myopathy.


Assuntos
Miopatias Distais/metabolismo , Miopatias Distais/patologia , Ácido N-Acetilneuramínico/deficiência , Estresse Oxidativo/fisiologia , Acetilcisteína/metabolismo , Acetilcisteína/uso terapêutico , Animais , Modelos Animais de Doenças , Humanos , Camundongos , Camundongos Knockout , Complexos Multienzimáticos/genética , Complexos Multienzimáticos/metabolismo , Proteínas Musculares/metabolismo , Músculo Esquelético/metabolismo , Atrofia Muscular/metabolismo , Atrofia Muscular/patologia , Ácido N-Acetilneuramínico/metabolismo , Espécies Reativas de Oxigênio/metabolismo
2.
PLoS One ; 12(3): e0173261, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28267778

RESUMO

GNE myopathy (GNEM), also known as hereditary inclusion body myopathy (HIBM), is a late- onset, progressive myopathy caused by mutations in the GNE gene encoding the enzyme responsible for the first regulated step in the biosynthesis of sialic acid (SA). The disease is characterized by distal muscle weakness in both the lower and upper extremities, with the quadriceps muscle relatively spared until the late stages of disease. To explore the role of SA synthesis in the disease, we conducted a comprehensive and systematic analysis of both free and total SA levels in a large cohort of GNEM patients and a mouse model. A sensitive LC/MS/MS assay was developed to quantify SA in serum and muscle homogenates. Mean serum free SA level was 0.166 µg/mL in patients and 18% lower (p<0.001) than that of age-matched control samples (0.203 µg/mL). In biopsies obtained from patients, mean free SA levels of different muscles ranged from 0.046-0.075 µg/µmol Cr and were markedly lower by 72-85% (p<0.001) than free SA from normal controls. Free SA was shown to constitute a small fraction (3-7%) of the total SA pool in muscle tissue. Differences in mean total SA levels in muscle from patients compared with normal controls were less distinct and more variable between different muscles, suggesting a small subset of sialylation targets could be responsible for the pathogenesis of GNEM. Normal quadriceps had significantly lower levels of free SA (reduced by 39%) and total SA (reduced by 53%) compared to normal gastrocnemius. A lower SA requirement for quadriceps may be linked to the reported quadriceps sparing in GNEM. Analysis of SA levels in GneM743T/M743T mutant mice corroborated the human study results. These results show that serum and muscle free SA is severely reduced in GNEM, which is consistent with the biochemical defect in SA synthesis associated with GNE mutations. These results therefore support the approach of reversing SA depletion as a potential treatment for GNEM patients.


Assuntos
Miopatias Distais/metabolismo , Músculo Esquelético/metabolismo , Ácido N-Acetilneuramínico/deficiência , Adolescente , Adulto , Idoso , Animais , Biomarcadores , Biópsia , Cromatografia Líquida , Modelos Animais de Doenças , Miopatias Distais/sangue , Miopatias Distais/patologia , Feminino , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Músculo Esquelético/patologia , Ácido N-Acetilneuramínico/sangue , Espectrometria de Massas em Tandem , Adulto Jovem
3.
Immunology ; 138(3): 235-45, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23113614

RESUMO

Dendritic cells (DCs) play an essential role in immunity against bacteria by phagocytosis and by eliciting adaptive immune responses. Previously, we demonstrated that human monocyte-derived DCs (MDDCs) express a high content of cell surface α2,6-sialylated glycans. However, the relative role of these sialylated structures in phagocytosis of bacteria has not been reported. Here, we show that treatment with a sialidase significantly improved the capacity of both immature and mature MDDCs to phagocytose Escherichia coli. Desialylated MDDCs had a significantly more mature phenotype, with higher expression of MHC molecules and interleukin (IL)-12, tumour necrosis factor-α, IL-6 and IL-10 cytokines, and nuclear factor-κB activation. T lymphocytes primed by desialylated MDDCs expressed more interferon-γ when compared with priming by sialylated MDDCs. Improved phagocytosis required E. coli sialic acids, indicating a mechanism of host-pathogen interaction dependent on sialic acid moieties. The DCs harvested from mice deficient in the ST6Gal.1 sialyltransferase showed improved phagocytosis capacity, demonstrating that the observed sialidase effect was a result of the removal of α2,6-sialic acid. The phagocytosis of different pathogenic E. coli isolates was also enhanced by sialidase, which suggests that modifications on MDDC sialic acids may be considered in the development of MDDC-based antibacterial therapies. Physiologically, our findings shed new light on mechanisms that modulate the function of both immature and mature MDDCs, in the context of host-bacteria interaction. Hence, with particular relevance to DC-based therapies, the engineering of α2,6-sialic acid cell surface is a novel possibility to fine tune DC phagocytosis and immunological potency.


Assuntos
Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Ácido N-Acetilneuramínico/deficiência , Fagocitose/imunologia , Animais , Células Cultivadas , Proteínas do Citoesqueleto/metabolismo , Escherichia coli/imunologia , Humanos , Camundongos , Camundongos Knockout , Ácido N-Acetilneuramínico/metabolismo , Fagocitose/genética , Proteínas rho de Ligação ao GTP/metabolismo
6.
Brain Nerve ; 62(6): 601-7, 2010 Jun.
Artigo em Japonês | MEDLINE | ID: mdl-20548120

RESUMO

Sialic acids are terminal sugars of glycolipids and glycoproteins and are involved in several cellular processes. Sialic acid biosynthesis occurs in the cytosol, where UDP-N-acetylglucosamine (GlcNAc) is sequentially converted to N-acetylmannosamine (ManNAc) 6-phosphate by UDP-GlcNAc-2-epimerase/ManNAc kinase enzymes, both of which are encoded by the GNE gene. Since the only existing mouse model of DMRV/hIBM (Gne(-/-)hGNED176VTg) exhibited decreased sialic acid levels in most organs, DMRV/hIBM is thought to be secondary to the metabolic defect in sialic acid production. Theoretically, replenishing sialic acid could be employed as a therapeutic option. It has been reported that N-acetylneuraminic acid (NeuAc) and ManNAc are well incorporated into cells and converted to sialic acid. Thus, we evaluated the efficacy and safety of ManNAc, NeuAc, and sialyllactose in the Gne(-/-)hGNED176VTg, by orally administering these agents to mice from 5-15 weeks continuously until they reached 54-57 weeks of age. The treatment showed beneficial effects in terms of survival rate, overall motor performance, myofiber size, ex vivo skeletal muscle contractile properties, and pathology. These low-dose compounds showed acceptable kidney and liver toxicity profiles. Thus our results show that the oral therapy with NeuAc and ManNAc or their derivatives is safe and effective in preventing myopathic symptoms in Gne(-/-)hGNED176VTg mice, and could be considered as a guide for further therapeutic trials.


Assuntos
Modelos Animais de Doenças , Miopatias Distais/tratamento farmacológico , Miopatias Distais/patologia , Hexosaminas/administração & dosagem , Corpos de Inclusão/patologia , Lactose/análogos & derivados , Camundongos Transgênicos , Ácido N-Acetilneuramínico/administração & dosagem , Ácidos Siálicos/administração & dosagem , Vacúolos/patologia , Administração Oral , Animais , Miopatias Distais/etiologia , Avaliação Pré-Clínica de Medicamentos , Humanos , Lactose/administração & dosagem , Camundongos , Ácido N-Acetilneuramínico/deficiência
8.
Rinsho Shinkeigaku ; 49(11): 852-5, 2009 Nov.
Artigo em Japonês | MEDLINE | ID: mdl-20030229

RESUMO

Distal myopathy with rimmed vacuoles (DMRV), also called hereditary inclusion body myopathy, is an autosomal recessive disorder caused by homozygous or compound heterozygous missense mutations in GNE which encodes a protein with two enzymatic activities in sialic acid biosynthesis: UDP-GlcNAc 2-epimerase and ManNAc kinase. The disease starts from 1540 years and is slowly progressive. DMRV preferentially affects tibialis anterior and hamstrings muscles, and has characteristic findings in muscle pathology which include rimmed vacuoles, tubulofilamentous inclusions, deposition of amyloid, and phosphorylated tau. We generated DMRV mice (Gne -/- hGNE D176V-Tg) by crossmating Gne knock-out heterozygous mouse and human GNE p.D176V transgenic mouse. This model mouse recapitulates DMRV clinically, pathologically, and biochemically by developing muscle weakness and atrophy from 21 weeks, amyloid deposition from 31 weeks, and rimmed vacuoles and phosphorylated tau from 41 weeks while having lifelong hyposialylation. We gave three types of GNE metabolites, ManNAc, NeuAc and sialyllactose, to DMRV mice orally from 15 weeks until 55 weeks of age. Sialic acid supplementation almost completely precluded the disease and virtually no sign of DMRV was seen even at 55 weeks of age, indicating that decreased sialic acid is the cause of myopathic phenotype and sialic acid supplementation can prevent the disease process.


Assuntos
Miopatias Distais/patologia , Miopatias Distais/terapia , Vacúolos/patologia , Amiloide/metabolismo , Animais , Atrofia , Miopatias Distais/etiologia , Humanos , Camundongos , Complexos Multienzimáticos/administração & dosagem , Complexos Multienzimáticos/genética , Músculo Esquelético/citologia , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Mutação de Sentido Incorreto , Ácido N-Acetilneuramínico/administração & dosagem , Ácido N-Acetilneuramínico/deficiência
9.
Eukaryot Cell ; 8(7): 1038-50, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19429777

RESUMO

Chitin is a globally abundant polymer widely distributed throughout eukaryotes that has been well characterized in only a few lineages. Diatoms are members of the eukaryotic lineage of stramenopiles. Of the hundreds of diatom genera, two produce long fibers of chitin that extrude through their cell walls of silica. We identify and describe here genes encoding putative chitin synthases in a variety of additional diatom genera, indicating that the ability to produce chitin is more widespread and likely plays a more central role in diatom biology than previously considered. Diatom chitin synthases fall into four phylogenetic clades. Protein domain predictions and differential gene expression patterns provide evidence that chitin synthases have multiple functions within a diatom cell. Thalassiosira pseudonana possesses six genes encoding three types of chitin synthases. Transcript abundance of the gene encoding one of these chitin synthase types increases when cells resume division after short-term silicic acid starvation and during short-term limitation by silicic acid or iron, two nutrient conditions connected in the environment and known to affect the cell wall. During long-term silicic acid starvation transcript abundance of this gene and one additional chitin synthase gene increased at the same time a chitin-binding lectin localized to the girdle band region of the cell wall. Together, these results suggest that the ability to produce chitin is more widespread in diatoms than previously thought and that a subset of the chitin produced by diatoms is associated with the cell wall.


Assuntos
Parede Celular/metabolismo , Quitina Sintase/metabolismo , Quitina/biossíntese , Diatomáceas/metabolismo , Parede Celular/química , Quitina/química , Quitina Sintase/classificação , Quitina Sintase/genética , Sequência Conservada/genética , Diatomáceas/química , Diatomáceas/genética , Evolução Molecular , Regulação da Expressão Gênica/genética , Lectinas/metabolismo , Ácido N-Acetilneuramínico/deficiência , Filogenia , Proteômica , Especificidade da Espécie
10.
Acta Myol ; 26(3): 171-5, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18646567

RESUMO

Distal myopathy with rimmed vacuoles (DMRV) or hereditary inclusion body myopathy (hIBM) is an adult onset slowly progressive myopathy secondary to mutations in the UDP-N-acetylglucosamine 2-epimerase/N-acetylmannosamine kinase (GNE) gene that encodes a bifunctional enzyme which catalyzes the rate-limiting step in sialic acid biosynthesis. Many hypotheses have been proposed to explain why patients develop weakness and atrophy, but are most views are obscure and thus are still considered controversial, partly because of the lack of an appropriate model with which these theories could be clarified. In this review, we briefly summarize the progress in DMRV research, and highlight efforts of researchers in generating the animal model for this myopathy.


Assuntos
Miopatias Distais , Músculo Esquelético/metabolismo , Ácido N-Acetilneuramínico/deficiência , Vacúolos/metabolismo , Animais , Modelos Animais de Doenças , Miopatias Distais/etiologia , Miopatias Distais/metabolismo , Miopatias Distais/patologia , Humanos , Músculo Esquelético/patologia , Vacúolos/patologia
11.
Int Immunol ; 18(4): 603-11, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16497829

RESUMO

CD22 is an inhibitory co-receptor of B cell receptor (BCR)-mediated signalling which binds specifically to glycan ligands containing alpha2,6-linked sialic acids. This interaction modulates the CD22 activity by an unknown mechanism. Mice deficient for ST6GalI, the enzyme that generates alpha2,6-linked sialic acids, show an immunodeficient and opposing phenotype to CD22-deficient mice. By generating mice double-deficient for this receptor/ligand pair, we analysed its influence on B cell maturation and signalling. Both ST6GalI-deficient and ST6GalI x CD22-deficient mice showed normal B cell development, but an impaired marginal zone B cell population in the spleen. Both types of mutant mice also showed a reduced population of bone marrow recirculating B cells, a defect previously detected in CD22-/- mice. In adoptive transfer experiments, a migration defect of wild-type B cells to the bone marrow of ST6GalI-deficient mice was found. This suggests a direct involvement of CD22 and its ligands 2,6Sia in a homing process of recirculating B cells to the bone marrow. Interestingly, defective B cell Ca2+ signalling and proliferation of ST6Gal-/- mice was rescued in ST6GalI x CD22-deficient mice. This points to a new mechanism of BCR signal regulation by CD22 and its ligand.


Assuntos
Linfócitos B/citologia , Linfócitos B/imunologia , Ácido N-Acetilneuramínico/imunologia , Lectina 2 Semelhante a Ig de Ligação ao Ácido Siálico/imunologia , Transdução de Sinais/imunologia , Transferência Adotiva , Animais , Movimento Celular , Proliferação de Células , Citometria de Fluxo , Ligantes , Ativação Linfocitária/imunologia , Camundongos , Camundongos Mutantes , Ácido N-Acetilneuramínico/deficiência , Receptores de Antígenos de Linfócitos B/imunologia , Sialiltransferases/deficiência , beta-D-Galactosídeo alfa 2-6-Sialiltransferase
12.
Prenat Diagn ; 24(3): 194-7, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15057952

RESUMO

BACKGROUND: Invasive trophoblast antigen (ITA), also known as hyperglycosylated hCG, is a glycosylation variant of hCG produced by invasive cytotrophoblast cells. Publications show that ITA is potentially a sensitive test for Down syndrome screening, reportedly detecting 38 and 80% of cases at a 5% false-positive rate in the first and second trimesters of pregnancy respectively. ITA has six oligosaccharides varying greatly in terminal sialic acid content (0-19 residues), leading to wide-charge heterogeneity. With the objective of developing an improved Down syndrome screening test, we examined the charge, or sialic acid variants, of ITA. METHODS: Twenty urine samples were collected from the 16th to 18th week of gestation from 10 patients with trisomy 21 fetuses, demonstrated by karyotype analysis, and from 10 patients with normal karyotype pregnancies. Proteins in each sample were separated by preparative isoelectric focusing. Fractions were collected, pooled into the pl ranges of 3.0 to 4.0, 4.1 to 5.0, 5.1 to 6.0 and 6.1 to 7.0 and assayed for ITA. RESULTS: A difference in the distribution of ITA charge variants was demonstrated between cases and controls, most apparent in the less acidic (pl 5.1-7.0) range. The controls and Down syndrome samples contained 26.7 +/- 14% versus 45.6 +/- 20%, respectively, of the less acidic form of ITA (P = 0.02). CONCLUSIONS: The ITA in Down syndrome cases is composed of a much higher proportion of less acidic ITA. This could be the major source of elevated levels of ITA in Down syndrome pregnancies. It is postulated that the development of a new assay specifically measuring ITA that is deficient in sialic acid might enhance the utility of the already highly sensitive screening tests for Down syndrome.


Assuntos
Gonadotropina Coriônica/urina , Síndrome de Down/diagnóstico , Programas de Rastreamento/métodos , Ácido N-Acetilneuramínico/deficiência , Trofoblastos/metabolismo , Biomarcadores/urina , Estudos de Casos e Controles , Gonadotropina Coriônica/metabolismo , Feminino , Idade Gestacional , Humanos , Focalização Isoelétrica , Cariotipagem , Neuraminidase/uso terapêutico , Gravidez , Primeiro Trimestre da Gravidez , Segundo Trimestre da Gravidez
13.
Am J Physiol Cell Physiol ; 281(2): C464-74, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11443045

RESUMO

We investigated the contribution of sialic acid residues to the K(+) currents involved in the repolarization of mouse ventricular myocytes. Ventricular K(+) currents had a rapidly inactivating component followed by slowly decaying and sustained components. This current was produced by the summation of three distinct currents: I(to), which contributed to the transient component; I(ss), which contributed to the sustained component; and I(K,slow), which contributed to both components. Incubation of ventricular myocytes with the sialidase neuraminidase reduced the amplitude of I(to) without altering I(K,slow) and I(ss). We found that the reduction in I(to) amplitude resulted from a depolarizing shift in the voltage of activation and a reduction in the conductance of I(to). Expression of Kv4.3 channels, a major contributor to I(to) in the ventricle, in a sialylation-deficient Chinese hamster ovary cell line (lec2) mimicked the effects of neuraminidase on the ventricular I(to). Furthermore, we showed that sialylated glycolipids have little effect on the voltage dependence of I(to). Finally, consistent with its actions on I(to), neuraminidase produced an increase in the duration of the action potential of ventricular myocytes and the frequency of early afterdepolarizations. We conclude that sialylation of the proteins forming Kv4 channels is important in determining the voltage dependence and conductance of I(to) and that incomplete glycosylation of these channels could lead to arrhythmias.


Assuntos
Ácido N-Acetilneuramínico/fisiologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana , Canais de Potássio/fisiologia , Função Ventricular , Potenciais de Ação/efeitos dos fármacos , Animais , Células CHO , Cálcio/farmacologia , Cricetinae , Condutividade Elétrica , Eletrofisiologia , Glicolipídeos/metabolismo , Homeostase , Miocárdio/citologia , Ácido N-Acetilneuramínico/deficiência , Neuraminidase/farmacologia , Canais de Potássio/efeitos dos fármacos , Tempo de Reação/efeitos dos fármacos , Canais de Potássio Shal
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA