Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 291
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
Basic Res Cardiol ; 115(6): 62, 2020 09 25.
Artigo em Inglês | MEDLINE | ID: mdl-32975669

RESUMO

Neuraminidase (NEU)1 forms a multienzyme complex with beta-galactosidase (ß-GAL) and protective-protein/cathepsin (PPC) A, which cleaves sialic-acids from cell surface glycoconjugates. We investigated the role of NEU1 in the myocardium after ischemia/reperfusion (I/R). Three days after inducing I/R, left ventricles (LV) of male mice (3 months-old) displayed upregulated neuraminidase activity and increased NEU1, ß-GAL and PPCA expression. Mice hypomorphic for neu1 (hNEU1) had less neuraminidase activity, fewer pro-inflammatory (Lin-CD11b+F4/80+Ly-6Chigh), and more anti-inflammatory macrophages (Lin-CD11b+F4/80+Ly-6Clow) 3 days after I/R, and less LV dysfunction 14 days after I/R. WT mice transplanted with hNEU1-bone marrow (BM) and hNEU1 mice with WT-BM showed significantly better LV function 14 days after I/R compared with WT mice with WT-BM. Mice with a cardiomyocyte-specific NEU1 overexpression displayed no difference in inflammation 3 days after I/R, but showed increased cardiomyocyte hypertrophy, reduced expression and mislocalization of Connexin-43 in gap junctions, and LV dysfunction despite a similar infarct scar size to WT mice 14 days after I/R. The upregulation of NEU1 after I/R contributes to heart failure by promoting inflammation in invading monocytes/macrophages, enhancing cardiomyocyte hypertrophy, and impairing gap junction function, suggesting that systemic NEU1 inhibition may reduce heart failure after I/R.


Assuntos
Insuficiência Cardíaca/etiologia , Hipertrofia Ventricular Esquerda/etiologia , Macrófagos/enzimologia , Monócitos/enzimologia , Infarto do Miocárdio/complicações , Traumatismo por Reperfusão Miocárdica/complicações , Miócitos Cardíacos/enzimologia , Neuraminidase/deficiência , Disfunção Ventricular Esquerda/etiologia , Animais , Catepsina A/metabolismo , Conexina 43/metabolismo , Modelos Animais de Doenças , Feminino , Junções Comunicantes/enzimologia , Junções Comunicantes/patologia , Insuficiência Cardíaca/enzimologia , Insuficiência Cardíaca/imunologia , Insuficiência Cardíaca/fisiopatologia , Hipertrofia Ventricular Esquerda/enzimologia , Hipertrofia Ventricular Esquerda/imunologia , Hipertrofia Ventricular Esquerda/fisiopatologia , Macrófagos/imunologia , Masculino , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Monócitos/imunologia , Infarto do Miocárdio/enzimologia , Infarto do Miocárdio/imunologia , Infarto do Miocárdio/patologia , Traumatismo por Reperfusão Miocárdica/enzimologia , Traumatismo por Reperfusão Miocárdica/imunologia , Traumatismo por Reperfusão Miocárdica/patologia , Miócitos Cardíacos/patologia , Neuraminidase/genética , Disfunção Ventricular Esquerda/enzimologia , Disfunção Ventricular Esquerda/imunologia , Disfunção Ventricular Esquerda/fisiopatologia , Função Ventricular Esquerda , Remodelação Ventricular , beta-Galactosidase/metabolismo
2.
J Virol ; 87(24): 13556-68, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24109212

RESUMO

Influenza viruses are highly genetically variable and escape from immunogenic pressure by antigenic changes in their surface proteins, referred to as "antigenic drift" and "antigenic shift." To assess the potential genetic plasticity under strong selection pressure, highly pathogenic avian influenza virus (HPAIV) of subtype H5N1 was passaged 50 times in embryonated chicken eggs in the presence of a neutralizing, polyclonal chicken serum. The resulting mutant acquired major alterations in the neuraminidase (NA)-encoding segment. Extensive deletions and rearrangements were detected, in contrast to only 12 amino acid substitutions within all other segments. Interestingly, this new neuraminidase segment resulted from complex sequence shuffling and insertion of a short fragment originating from the PA segment. Characterization of that novel variant revealed a loss of the neuraminidase protein and enzymatic activity, but its replication efficiency remained comparable to that of the wild type. Using reverse genetics, a recombinant virus consisting of the wild-type backbone and the shortened NA segment could be generated; however, generation of this recombinant virus required the polybasic hemagglutinin cleavage site. Two independent repetitions starting with egg passage 30 in the presence of alternative chicken-derived immune sera selected mutants with similar but different large deletions within the NA segment without any neuraminidase activity, indicating a general mechanism. In chicken, these virus variants were avirulent, even though the HPAIV polybasic hemagglutinin cleavage site was still present. Overall, the variants reported here are the first HPAIV H5N1 strains without a functional neuraminidase shown to grow efficiently without any helper factor. These novel HPAIV variants may facilitate future studies shedding light on the role of neuraminidase in virus replication and pathogenicity.


Assuntos
Virus da Influenza A Subtipo H5N1/enzimologia , Virus da Influenza A Subtipo H5N1/fisiologia , Influenza Aviária/virologia , Neuraminidase/genética , Doenças das Aves Domésticas/virologia , Deleção de Sequência , Proteínas Virais/genética , Replicação Viral , Substituição de Aminoácidos , Animais , Galinhas , Virus da Influenza A Subtipo H5N1/genética , Mutagênese Insercional , Neuraminidase/química , Neuraminidase/deficiência , Inoculações Seriadas , Proteínas Virais/química , Proteínas Virais/metabolismo
3.
Arch Virol ; 159(4): 797-800, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24142272

RESUMO

We identified a novel neuraminidase (NA)-deficient virus that was a 2009 pandemic influenza H1N1 virus mutant. The mutant virus had a deletion of 1,009 nt in the NA gene and lacked an enzymatic domain. Although the yield of the NA-deficient virus was limited, it formed large plaques when applied to MDCK cell cultures, indicating that the virus was able to spread to adjacent cells. Furthermore, the NA-deficient virus was eluted from chicken erythrocytes at 37 °C, even in the presence of the antiviral drug peramivir. Spread of this NA-deficient virus may pose a potential threat to anti-influenza therapies.


Assuntos
Vírus da Influenza A Subtipo H1N1/fisiologia , Neuraminidase/deficiência , Replicação Viral , Animais , Linhagem Celular , Galinhas , Cães , Humanos , Vírus da Influenza A Subtipo H1N1/genética , Vírus da Influenza A Subtipo H1N1/isolamento & purificação , Influenza Humana/virologia , Dados de Sequência Molecular , RNA Viral/genética , Análise de Sequência de DNA , Deleção de Sequência , Ensaio de Placa Viral , Proteínas Virais
4.
Biochemistry (Mosc) ; 78(7): 736-45, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24010837

RESUMO

Terminal sialic acid residues are found in abundance in glycan chains of glycoproteins and glycolipids on the surface of all live cells forming an outer layer of the cell originally known as glycocalyx. Their presence affects the molecular properties and structure of glycoconjugates, modifying their function and interactions with other molecules. Consequently, the sialylation state of glycoproteins and glycolipids has been recognized as a critical factor modulating molecular recognitions inside the cell, between the cells, between the cells and the extracellular matrix, and between the cells and certain exogenous pathogens. Until recently sialyltransferases that catalyze transfer of sialic acid residues to the glycan chains in the process of their biosynthesis were thought to be mainly responsible for the creation and maintenance of a temporal and spatial diversity of sialylated moieties. However, the growing evidence suggests that in mammalian cells, at least equally important roles belong to sialidases/neuraminidases, which are located on the cell surface and in intracellular compartments, and may either initiate the catabolism of sialoglycoconjugates or just cleave their sialic acid residues, and thereby contribute to temporal changes in their structure and functions. The current review summarizes emerging data demonstrating that mammalian neuraminidase 1, well known for its lysosomal catabolic function, is also targeted to the cell surface and assumes the previously unrecognized role as a structural and functional modulator of cellular receptors.


Assuntos
Ácidos Siálicos/metabolismo , Transdução de Sinais , Animais , Glicocálix/química , Glicocálix/metabolismo , Glicoconjugados/química , Glicoconjugados/metabolismo , Glicolipídeos/química , Glicolipídeos/metabolismo , Glicoproteínas/química , Glicoproteínas/metabolismo , Humanos , Linfócitos/imunologia , Linfócitos/metabolismo , Proteína 1 de Membrana Associada ao Lisossomo/metabolismo , Neuraminidase/deficiência , Neuraminidase/genética , Neuraminidase/metabolismo , Ácidos Siálicos/química , Sialiltransferases/metabolismo
5.
PLoS Genet ; 6(9): e1001118, 2010 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-20862357

RESUMO

Tay-Sachs disease is a severe lysosomal disorder caused by mutations in the HexA gene coding for the α-subunit of lysosomal ß-hexosaminidase A, which converts G(M2) to G(M3) ganglioside. Hexa(-/-) mice, depleted of ß-hexosaminidase A, remain asymptomatic to 1 year of age, because they catabolise G(M2) ganglioside via a lysosomal sialidase into glycolipid G(A2), which is further processed by ß-hexosaminidase B to lactosyl-ceramide, thereby bypassing the ß-hexosaminidase A defect. Since this bypass is not effective in humans, infantile Tay-Sachs disease is fatal in the first years of life. Previously, we identified a novel ganglioside metabolizing sialidase, Neu4, abundantly expressed in mouse brain neurons. Now we demonstrate that mice with targeted disruption of both Neu4 and Hexa genes (Neu4(-/-);Hexa(-/-)) show epileptic seizures with 40% penetrance correlating with polyspike discharges on the cortical electrodes of the electroencephalogram. Single knockout Hexa(-/-) or Neu4(-/-) siblings do not show such symptoms. Further, double-knockout but not single-knockout mice have multiple degenerating neurons in the cortex and hippocampus and multiple layers of cortical neurons accumulating G(M2) ganglioside. Together, our data suggest that the Neu4 block exacerbates the disease in Hexa(-/-) mice, indicating that Neu4 is a modifier gene in the mouse model of Tay-Sachs disease, reducing the disease severity through the metabolic bypass. However, while disease severity in the double mutant is increased, it is not profound suggesting that Neu4 is not the only sialidase contributing to the metabolic bypass in Hexa(-/-) mice.


Assuntos
Epilepsia/enzimologia , Epilepsia/patologia , Lisossomos/enzimologia , Neuraminidase/deficiência , Neurônios/enzimologia , Neurônios/patologia , Cadeia alfa da beta-Hexosaminidase/metabolismo , Animais , Comportamento Animal , Córtex Cerebral/enzimologia , Córtex Cerebral/patologia , Córtex Cerebral/fisiopatologia , Córtex Cerebral/ultraestrutura , Eletroencefalografia , Epilepsia/fisiopatologia , Gangliosídeo G(M2)/metabolismo , Técnicas de Inativação de Genes , Hipocampo/enzimologia , Hipocampo/patologia , Hipocampo/fisiopatologia , Hipocampo/ultraestrutura , Aprendizagem/fisiologia , Lisossomos/patologia , Lisossomos/ultraestrutura , Camundongos , Atividade Motora/fisiologia , Neuraminidase/metabolismo , Neurônios/ultraestrutura
6.
Nat Genet ; 15(3): 316-20, 1997 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-9054950

RESUMO

Sialidase (neuraminidase, EC 3.2.1.18) catalyses the hydrolysis of terminal sialic acid residues of glyconjugates. Sialidase has been well studied in viruses and bacteria where it destroys the sialic acid-containing receptors at the surface of host cells, and mobilizes bacterial nutrients. In mammals, three types of sialidases, lysosomal, plasma membrane and cytosolic, have been described. For lysosomal sialidase in humans, the primary genetic deficiency results in an autosomal recessive disease, sialidosis, associated with tissue accumulation and urinary excretion of sialylated oligosaccharides and glycolipids. Sialidosis includes two main clinical variants: late-onset, sialidosis type I, characterized by bilateral macular cherry-red spots and myoclonus, and infantile-onset, sialidosis type II, characterized by skeletal dysplasia, mental retardation and hepatosplenomegaly. We report the identification of human lysosomal sialidase cDNA, its cloning, sequencing and expression. Examination of six sialidosis patients revealed three mutations, one frameshift insertion and two missense. We mapped the lysosomal sialidase gene to human chromosome 6 (6p21.3), which is consistent with the previous chromosomal assignment of this gene in proximity to the HLA locus.


Assuntos
Cromossomos Humanos Par 6 , Doenças por Armazenamento dos Lisossomos/enzimologia , Doenças por Armazenamento dos Lisossomos/genética , Mutação , Neuraminidase/genética , Sequência de Aminoácidos , Sequência de Bases , Células Cultivadas , Mapeamento Cromossômico , Clonagem Molecular , Primers do DNA , Elementos de DNA Transponíveis , Mutação da Fase de Leitura , Humanos , Lisossomos/enzimologia , Dados de Sequência Molecular , Neuraminidase/química , Neuraminidase/deficiência , Mutação Puntual , Reação em Cadeia da Polimerase , Polimorfismo Conformacional de Fita Simples , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/química , Homologia de Sequência de Aminoácidos , Pele/enzimologia
7.
J Biol Chem ; 285(1): 206-15, 2010 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-19889639

RESUMO

The differentiation of monocytes into macrophages and dendritic cells is accompanied by induction of cell-surface neuraminidase 1 (Neu1) and cathepsin A (CathA), the latter forming a complex with and activating Neu1. To clarify the biological importance of this phenomenon we have developed the gene-targeted mouse models of a CathA deficiency (CathA(S190A)) and a double CathA/Neu1 deficiency (CathA(S190A-Neo)). Macrophages of CathA(S190A-Neo) mice and their immature dendritic cells showed a significantly reduced capacity to engulf Gram-positive and Gram-negative bacteria and positively and negatively charged polymer beads as well as IgG-opsonized beads and erythrocytes. Properties of the cells derived from CathA(S190A) mice were indistinguishable from those of wild-type controls, suggesting that the absence of Neu1, which results in the increased sialylation of the cell surface proteins, probably affects multiple receptors for phagocytosis. Indeed, treatment of the cells with purified mouse Neu1 reduced surface sialylation and restored phagocytosis. Because Neu1-deficient cells showed reduced internalization of IgG-opsonized sheep erythrocytes whereas binding of the erythrocytes to the cells at 4 degrees C persisted, we speculate that the absence of Neu1 in particular affected transduction of signals from the Fc receptors for immunoglobulin G (FcgammaR). Indeed the macrophages from the Neu1-deficient mice showed increased sialylation and impaired phosphorylation of FcgammaR as well as markedly reduced phosphorylation of Syk kinase in response to treatment with IgG-opsonized beads. Altogether our data suggest that the cell surface Neu1 activates the phagocytosis in macrophages and dendritic cells through desialylation of surface receptors, thus, contributing to their functional integrity.


Assuntos
Macrófagos/citologia , Macrófagos/enzimologia , Neuraminidase/metabolismo , Fagocitose , Animais , Catepsina A/metabolismo , Diferenciação Celular , Membrana Celular/enzimologia , Células Dendríticas/citologia , Eritrócitos/citologia , Eritrócitos/metabolismo , Lectinas/metabolismo , Macrófagos/microbiologia , Camundongos , Ácido N-Acetilneuramínico/metabolismo , Neuraminidase/deficiência , Proteínas Opsonizantes/imunologia , Receptores de IgG/imunologia , Ovinos , Transdução de Sinais , Coloração e Rotulagem
8.
Biochim Biophys Acta ; 1802(2): 259-68, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19857571

RESUMO

The neuraminidase-1 (Neu1) knockout mouse model is a phenocopy of the lysosomal storage disease (LSD) sialidosis, characterized by multisystemic and neuropathic symptoms, including hearing loss. We have characterized the auditory defects in Neu1(-/-) mice and found that hearing loss involves both conductive and sensorineural components. Auditory brainstem response (ABR) thresholds were significantly elevated in Neu1(-/-) mice at P21 (48-55 dB), and hearing loss appeared progressive (53-66 dB at P60). At these ages Neu1(-/-) mice accumulated cerumen in the external ear canal and had a thickened mucosa and inflammation in the middle ear. In cochleae of adult wild-type mice, Neu1 was expressed in several cell types in the stria vascularis, the organ of Corti, and spiral ganglion. Progressive morphological abnormalities such as extensive vacuolization were detected in the Neu1(-/-) cochleae as early as P9. These early morphologic changes in Neu1(-/-) cochleae were associated with oversialylation of several lysosomal associated membrane proteins (Lamps) in the stria vascularis. A marked increase in the expression and apical localization of Lamp-1 in marginal cells of the stria vascularis predicts exacerbation of lysosomal exocytosis into the endolymph. Consequently, the endolymphatic potential in Neu1(-/-) mice was reduced by approximately 20 mV at ages P31-P44, which would cause dysfunction of transduction in sensory hair cells. This study suggests a molecular mechanism that contributes to hearing loss in sialidosis and identifies potential therapeutic targets.


Assuntos
Cóclea/fisiopatologia , Perda Auditiva/genética , Neuraminidase/deficiência , Neuraminidase/genética , Animais , Cóclea/patologia , Modelos Animais de Doenças , Orelha Média/patologia , Perda Auditiva/enzimologia , Doenças por Armazenamento dos Lisossomos/enzimologia , Doenças por Armazenamento dos Lisossomos/genética , Camundongos , Camundongos Knockout , Microscopia Eletrônica de Varredura , Vacúolos/patologia , Vacúolos/ultraestrutura
9.
Biochim Biophys Acta ; 1802(7-8): 659-72, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20388541

RESUMO

Neuraminidase 1 (NEU1) regulates the catabolism of sialoglycoconjugates in lysosomes. Congenital NEU1 deficiency in children is the basis of sialidosis, a severe neurosomatic disorder in which patients experience a broad spectrum of clinical manifestations varying in the age of onset and severity. Osteoskeletal deformities and muscle hypotonia have been described in patients with sialidosis. Here we present the first comprehensive analysis of the skeletal muscle pathology associated with loss of Neu1 function in mice. In this animal model, skeletal muscles showed an expansion of the epimysial and perimysial spaces, associated with proliferation of fibroblast-like cells and abnormal deposition of collagens. Muscle fibers located adjacent to the expanded connective tissue underwent extensive invagination of their sarcolemma, which resulted in the infiltration of the fibers by fibroblast-like cells and extracellular matrix, and in their progressive cytosolic fragmentation. Both the expanded connective tissue and the juxtaposed infiltrated muscle fibers were strongly positive for lysosomal markers and displayed increased proteolytic activity of lysosomal cathepsins and metalloproteinases. These combined features could lead to abnormal remodeling of the extracellular matrix that could be responsible for sarcolemmal invagination and progressive muscle fiber degeneration, ultimately resulting in an overt atrophic phenotype. This unique pattern of muscle damage, which has never been described in any myopathy, might explain the neuromuscular manifestations reported in patients with the type II severe form of sialidosis. More broadly, these findings point to a potential role of NEU1 in cell proliferation and extracellular matrix remodeling.


Assuntos
Tecido Conjuntivo/fisiopatologia , Fibras Musculares Esqueléticas/patologia , Atrofia Muscular/etiologia , Atrofia Muscular/patologia , Neuraminidase/genética , Animais , Apoptose/genética , Apoptose/fisiologia , Movimento Celular/genética , Proliferação de Células , Tecido Conjuntivo/patologia , Matriz Extracelular/patologia , Matriz Extracelular/fisiologia , Fibroblastos/patologia , Camundongos , Camundongos Knockout , Atrofia Muscular/genética , Atrofia Muscular/fisiopatologia , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/patologia , Necrose/genética , Necrose/patologia , Neuraminidase/deficiência , Neuraminidase/fisiologia , Sarcolema/patologia
10.
Microb Pathog ; 50(6): 343-9, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21377521

RESUMO

Pneumococcal hemolytic uremic syndrome is recognized in a small portion of otherwise healthy children who have or have recently had Streptococcus pneumoniae infections, including severe pneumonia, meningitis, and bacteremia. As in other types of hemolytic uremic syndrome (HUS), pneumococcal HUS is characterized by microangiopathic hemolytic anemia, and thrombocytopenia, usually with extensive kidney damage. Although not demonstrated in vivo, the pathogenesis of pneumococcal HUS has been attributed to the action pneumococcal neuraminidase exposing the usually cryptic Thomsen-Friedenreich antigen (T-antigen) on red blood cells (RBC), and kidney glomeruli. We evaluated the effect of pneumococcal infection on desialylation of RBC and glomeruli during pneumococcal infections in mice. Following intravenous infection with capsular type 19F pneumococci, CFU levels exceeding 1000 CFU/mL blood by the third day were significantly more likely to result in exposed T-antigen on RBC than lower levels of bacteremia. In a pneumonia model, significantly more T-antigen was exposed on RBC in mice treated with penicillin than in those receiving mock treatment. Utilizing mutant pneumococci, we demonstrated that neuraminidase A but not neuraminidase B was necessary for exposure of T-antigen on RBC in vivo. Thus, pneumococcal neuraminidase A is necessary for the exposure of T-antigen in vivo and treatment with penicillin increases this effect. Interestingly, NanA(-) pneumococci were found in the blood in higher numbers and caused more deaths than wild type, NanB(-), or the NanA(-)/NanB(-) pneumococci.


Assuntos
Antígenos Glicosídicos Associados a Tumores/imunologia , Neuraminidase/imunologia , Infecções Pneumocócicas/imunologia , Streptococcus pneumoniae/imunologia , Animais , Antígenos Virais de Tumores/sangue , Antígenos Virais de Tumores/imunologia , Eritrócitos/imunologia , Feminino , Síndrome Hemolítico-Urêmica/sangue , Síndrome Hemolítico-Urêmica/imunologia , Síndrome Hemolítico-Urêmica/microbiologia , Rim/enzimologia , Rim/imunologia , Meningite Pneumocócica/sangue , Meningite Pneumocócica/imunologia , Meningite Pneumocócica/microbiologia , Camundongos , Camundongos Endogâmicos CBA , Neuraminidase/deficiência , Infecções Pneumocócicas/sangue , Infecções Pneumocócicas/microbiologia , Pneumonia Bacteriana/sangue , Pneumonia Bacteriana/imunologia , Pneumonia Bacteriana/microbiologia , Streptococcus pneumoniae/enzimologia
11.
Ultrastruct Pathol ; 35(4): 168-71, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21568622

RESUMO

Nephrosialidosis is a rare subgroup of sialidosis characterized by a progressive and fatal course of nephropathy. The authors report a 2-year-old boy who had suffered from steroid-resistant nephrotic syndrome. Renal biopsy showed diffuse and severe vacuolization of glomerular and tubular epithelial cells. The vacuoles were ultrastructurally membrane bound, most of which were not empty but contained electron-dense material lining the inner surface of the membrane. The pathologic changes were consistent with neuraminidase-deficient disorders, which was later confirmed by biochemical analysis. Although rare, nephrosialidosis should be considered in children with steroid-resistant nephrotic syndrome if renal biopsies exhibit severe vacuolar degeneration of renal epithelial cells.


Assuntos
Rim/ultraestrutura , Mucolipidoses/patologia , Síndrome Nefrótica/congênito , Biópsia , Encefalopatias Metabólicas/patologia , Encefalopatias Metabólicas/fisiopatologia , Pré-Escolar , Evolução Fatal , Humanos , Falência Renal Crônica , Masculino , Microscopia Eletrônica de Transmissão , Mucolipidoses/complicações , Síndrome Nefrótica/complicações , Síndrome Nefrótica/patologia , Neuraminidase/deficiência
12.
Sci Rep ; 11(1): 13477, 2021 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-34188220

RESUMO

NEU1 sialidase hydrolyzes sialic acids from glycoconjugates in lysosomes. Deficiency of NEU1 causes sialidosis with symptoms including facial dysmorphism, bone dysplasia, and neurodegeneration. However, the effects of NEU1 deficiency on emotional activity have not been explored. Here, we conducted the behavioral analysis using Neu1-knockout zebrafish (Neu1-KO). Neu1-KO zebrafish showed normal swimming similar to wild-type zebrafish (WT), whereas shoaling was decreased and accompanied by greater inter-fish distance than WT zebrafish. The aggression test showed a reduced aggressive behavior in Neu1-KO zebrafish than in WT zebrafish. In the mirror and 3-chambers test, Neu1-KO zebrafish showed more interest toward the opponent in the mirror and multiple unfamiliar zebrafish, respectively, than WT zebrafish. Furthermore, Neu1-KO zebrafish also showed increased interaction with different fish species, whereas WT zebrafish avoided them. In the black-white preference test, Neu1-KO zebrafish showed an abnormal preference for the white region, whereas WT zebrafish preferred the black region. Neu1-KO zebrafish were characterized by a downregulation of the anxiety-related genes of the hypothalamic-pituitary-adrenal axis and upregulation of lamp1a, an activator of lysosomal exocytosis, with their brains accumulating several sphingoglycolipids. This study revealed that Neu1 deficiency caused abnormal emotional behavior in zebrafish, possibly due to neuronal dysfunction induced by lysosomal exocytosis.


Assuntos
Comportamento Animal , Emoções , Neuraminidase/deficiência , Comportamento Social , Proteínas de Peixe-Zebra/deficiência , Peixe-Zebra , Animais , Técnicas de Inativação de Genes , Neuraminidase/metabolismo , Proteínas de Peixe-Zebra/metabolismo
13.
J Gen Virol ; 91(Pt 11): 2753-61, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20702651

RESUMO

Proteolytic cleavage of haemagglutinin (HA) is essential for the infectivity of influenza A viruses (IAVs). This is usually mediated by trypsin-like proteases present in the respiratory tract. However, the ability to use plasminogen (PLG) as an alternative protease may contribute to pathogenesis of IAV infections and virus replication outside the respiratory tract. It was demonstrated previously that neuraminidase (NA) of the IAV strain A/WSN/33 can sequester PLG, allowing this virus to replicate in a PLG-dependent fashion. However, PLG also promotes replication of other IAVs, although its mode of action is poorly understood. Here, using NA-deficient viruses, we demonstrate that NA is not required for the binding of PLG and subsequent cleavage of HA. However, we demonstrate that the cellular protein annexin 2 (A2) can bind PLG and contributes to PLG-dependent cleavage of HA and subsequent IAV replication. Collectively, these results indicate that PLG promotes IAV replication in an A2-dependent fashion in the absence of NA.


Assuntos
Anexina A2/metabolismo , Interações Hospedeiro-Patógeno , Vírus da Influenza A/fisiologia , Neuraminidase/deficiência , Plasminogênio/metabolismo , Internalização do Vírus , Replicação Viral , Animais , Linhagem Celular , Humanos , Carga Viral , Proteínas Virais
14.
Mol Genet Metab ; 97(1): 43-52, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19217813

RESUMO

SM/J is an inbred mouse strain with a complex phenotype including small body size, impaired immune response and a tissue-specific sialidase deficiency. We identified a regulatory mutation, (-519G-->A) within the neu1 promoter which in reporter assays resulted in significantly reduced transcription. This mutation generates a consensus binding site for Nkx3 family transcription repressors. Recombinant Nkx3.2 bound strongly to and preferentially repressed transcription of the mutant promoter. This tissue-specific deficiency results in a retarded immune response and modulates leukocyte recruitment. Examination of the hepatic microcirculation in mutant mice revealed increased rolling and decreased adhesion of leukocytes. Our findings support a significant role for lysosomal sialidase in inflammation and highlight the significance of repressor-recruitment in genetic disease.


Assuntos
Proteínas de Homeodomínio/metabolismo , Neuraminidase/deficiência , Neuraminidase/genética , Mutação Puntual/genética , Regiões Promotoras Genéticas , Proteínas Repressoras/metabolismo , Fatores de Transcrição/metabolismo , Animais , Sequência de Bases , Linhagem Celular , Movimento Celular , DNA/metabolismo , Modelos Animais de Doenças , Ensaio de Desvio de Mobilidade Eletroforética , Inflamação/enzimologia , Leucócitos/citologia , Fígado/irrigação sanguínea , Fígado/citologia , Camundongos , Microcirculação , Dados de Sequência Molecular , Especificidade de Órgãos , Ligação Proteica , Transcrição Gênica
15.
Glycoconj J ; 26(9): 1197-212, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19430901

RESUMO

The signaling pathways of mammalian Toll-like receptors (TLR) are well characterized, but the initial molecular mechanisms activated following ligand interactions with the receptors remain poorly defined. Here, we show a membrane controlling mechanism that is initiated by ligand binding to TLR-2, -3 and-4 to induce Neu1 sialidase activity within minutes in live primary bone marrow (BM) macrophage cells and macrophage and dendritic cell lines. Central to this process is that Neu1 and not Neu2,-3 and-4 forms a complex with TLR-2,-3 and-4 on the cell surface of naïve macrophage cells. Neuraminidase inhibitors BCX1827, 2-deoxy-2,3-dehydro-N-acetylneuraminic acid (DANA), zanamivir and oseltamivir carboxylate have a limited significant inhibition of the LPS-induced sialidase activity in live BMC-2 macrophage cells but Tamiflu (oseltamivir phosphate) completely blocks this activity. Tamiflu inhibits LPS-induced sialidase activity in live BMC-2 cells with an IC(50) of 1.2 microM compared to an IC(50) of 1015 microM for its hydrolytic metabolite oseltamivir carboxylate. Tamiflu blockage of LPS-induced Neu1 sialidase activity is not affected in BMC-2 cells pretreated with anticarboxylesterase agent clopidogrel. Endotoxin LPS binding to TLR4 induces Neu1 with subsequent activation of NFkappaB and the production of nitric oxide and pro-inflammatory IL-6 and TNFalpha cytokines in primary and macrophage cell lines. Hypomorphic cathepsin A mice with a secondary Neu1 deficiency respond poorly to LPS-induced pro-inflammatory cytokines compared to the wild-type or hypomorphic cathepsin A with normal Neu1 mice. Our findings establish an unprecedented mechanism for pathogen molecule-induced TLR activation and cell function, which is critically dependent on Neu1 sialidase activity associated with TLR ligand treated live primary macrophage cells and macrophage and dendritic cell lines.


Assuntos
Células Dendríticas/enzimologia , Macrófagos/enzimologia , Neuraminidase/metabolismo , Receptores de Reconhecimento de Padrão/metabolismo , Receptores Toll-Like/imunologia , Animais , Células da Medula Óssea/citologia , Membrana Celular/efeitos dos fármacos , Membrana Celular/enzimologia , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Citocinas/sangue , Células Dendríticas/citologia , Células Dendríticas/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Humanos , Imunoprecipitação , Mediadores da Inflamação/metabolismo , Ligantes , Lipopolissacarídeos/farmacologia , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Camundongos , Neuraminidase/antagonistas & inibidores , Neuraminidase/deficiência , Óxido Nítrico/biossíntese , Oseltamivir/farmacologia
16.
Brain Dev ; 30(9): 595-8, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18396002

RESUMO

Galactosialidosis is a rare lysosomal storage disease caused by a combined deficiency of lysosomal beta-galactosidase and neuraminidase, due to a primary defect in protective protein/cathepsin A. Three subtypes are recognized: the early infantile type, the late infantile type, and the juvenile/adult type. Here, we report a case of early infantile galactosialidosis in a female who was born at 31 weeks of gestation, after detection of fetal ascites at 21 weeks of gestation and development of fetal hydrops. After birth she received intensive treatment that led to improvement of edema and pleural effusion, but ascites slowly developed. She died of renal failure on day 207. An autopsy showed that all organs contained vacuolated cells, compatible with a storage disease. The patient had decreased activity of beta-galactosidase and undetectable neuraminidase activity in fibroblasts. A single A-G base transition at position 146 of exon 1 (Q49R) in protective protein/cathepsin A gene was found. The mutation has been reported previously in a Japanese patient with different phenotypes. However homozygous Q49R mutation detected in our case was severe prognosis.


Assuntos
Catepsina A/genética , Doenças por Armazenamento dos Lisossomos/genética , Neuraminidase/deficiência , Mutação Puntual , beta-Galactosidase/deficiência , Encéfalo/metabolismo , Encéfalo/patologia , Catepsina A/metabolismo , Análise Mutacional de DNA , Evolução Fatal , Feminino , Humanos , Hidropisia Fetal/etiologia , Hidropisia Fetal/patologia , Lactente , Recém-Nascido , Rim/metabolismo , Rim/patologia , Fígado/metabolismo , Fígado/patologia , Doenças por Armazenamento dos Lisossomos/complicações , Doenças por Armazenamento dos Lisossomos/metabolismo , Doenças por Armazenamento dos Lisossomos/patologia , Miocárdio/metabolismo , Miocárdio/patologia
17.
Exp Neurol ; 299(Pt A): 26-41, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28974375

RESUMO

Tay-Sachs disease is a severe lysosomal storage disorder caused by mutations in Hexa, the gene that encodes for the α subunit of lysosomal ß-hexosaminidase A (HEXA), which converts GM2 to GM3 ganglioside. Unexpectedly, Hexa-/- mice have a normal lifespan and show no obvious neurological impairment until at least one year of age. These mice catabolize stored GM2 ganglioside using sialidase(s) to remove sialic acid and form the glycolipid GA2, which is further processed by ß-hexosaminidase B. Therefore, the presence of the sialidase (s) allows the consequences of the Hexa defect to be bypassed. To determine if the sialidase NEU3 contributes to GM2 ganglioside degradation, we generated a mouse model with combined deficiencies of HEXA and NEU3. The Hexa-/-Neu3-/- mice were healthy at birth, but died at 1.5 to 4.5months of age. Thin-layer chromatography and mass spectrometric analysis of the brains of Hexa-/-Neu3-/- mice revealed the abnormal accumulation of GM2 ganglioside. Histological and immunohistochemical analysis demonstrated cytoplasmic vacuolation in the neurons. Electron microscopic examination of the brain, kidneys and testes revealed pleomorphic inclusions of many small vesicles and complex lamellar structures. The Hexa-/-Neu3-/- mice exhibited progressive neurodegeneration with neuronal loss, Purkinje cell depletion, and astrogliosis. Slow movement, ataxia, and tremors were the prominent neurological abnormalities observed in these mice. Furthermore, radiographs revealed abnormalities in the skeletal bones of the Hexa-/-Neu3-/- mice. Thus, the Hexa-/-Neu3-/- mice mimic the neuropathological and clinical abnormalities of the classical early-onset Tay-Sachs patients, and provide a suitable model for the future pre-clinical testing of potential treatments for this condition.


Assuntos
Gangliosidoses GM2/genética , Hexosaminidase B/genética , Neuraminidase/genética , Doença de Tay-Sachs/genética , Animais , Química Encefálica/genética , Vesículas Citoplasmáticas/patologia , Gangliosidoses GM2/metabolismo , Gliose/genética , Gliose/patologia , Glicoesfingolipídeos/metabolismo , Coxeadura Animal/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neuraminidase/deficiência , Neurônios/patologia , Células de Purkinje/patologia , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Doença de Tay-Sachs/patologia
18.
Artigo em Inglês | MEDLINE | ID: mdl-29675399

RESUMO

Porphyromonas gingivalis (P. gingivalis) is a major periodontal pathogen that can induce an immune response leading to a destructive inflammatory process. During the inflammatory process, interleukin-12 (IL-12) is secreted, correlating with bacterial clearance by macrophages. Bacterial sialidase has recently been shown to influence the synthesis and modification of the macromolecules on its surface, and is associated with the interaction between bacteria and host cells. We have previously constructed a P. gingivalis sialidase gene mutant strain in P. gingivalis W83 (ΔPG0352) and found that ΔPG0352 showed less pathogenicity than the wild-type strain. In this study, U937-differentiated macrophages were stimulated by P. gingivalis W83, ΔPG0352, or PG0352 complemented strain (comΔPG0352). Transmission electron microscopy showed that P. gingivalis caused a loss of membrane integrity in macrophages and the intracellular bacteria were enclosed within endocytic vacuoles. The expression of both IL-12p35 and IL-12p40 genes and the levels of IL-12p70 were significantly higher in U937 stimulated by ΔPG0352 than in those with P. gingivalis W83 and comΔPG0352. In order to explain why ΔPG0352 induced more IL-12 in macrophages, immunofluorescence assays, PCR arrays, and gene silence or overexpression experiments were carried out. Immunofluorescence assays showed that ΔPG0352 induced lower expression of CR3 in macrophages. After CR3 was suppressed, there were no significant differences in the IL-12p70 levels between macrophages stimulated by P. gingivalis W83, ΔPG0352 or comΔPG0352. PCR array experiments showed that miR-21 and lncRNA GAS5 were differentially expressed between macrophages stimulated by P. gingivalis W83 and ΔPG0352, which had been identified by real-time PCR. The results of CR3 blocking and lncRNA GAS5 gene silence or overexpression showed that the difference in IL-12 levels between P. gingivalis W83 and ΔPG0352 groups was associated with CR3, lncRNA GAS5 and miR-21. Thus it can be concluded that the sialidase-deficient strain is more easily cleared by attenuating CR3 activation, reducing the inhibition of lncRNA GAS5, inducing less miR-21 and more IL-12 in macrophages. These results indicate that inhibiting the activity of sialidase in P. gingivalis will cause rapid clearing by macrophages.


Assuntos
Proteínas de Bactérias/metabolismo , Infecções por Bacteroidaceae/metabolismo , Interleucina-12/metabolismo , Antígeno de Macrófago 1/metabolismo , MicroRNAs/metabolismo , Neuraminidase/deficiência , Porphyromonas gingivalis/enzimologia , RNA Longo não Codificante/metabolismo , Proteínas de Bactérias/genética , Infecções por Bacteroidaceae/genética , Infecções por Bacteroidaceae/microbiologia , Linhagem Celular , Interações Hospedeiro-Patógeno , Humanos , Interleucina-12/genética , Antígeno de Macrófago 1/genética , Macrófagos/metabolismo , MicroRNAs/genética , Neuraminidase/genética , Porphyromonas gingivalis/genética , RNA Longo não Codificante/genética
20.
J Biochem ; 162(2): 137-143, 2017 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-28130415

RESUMO

A sialidase NEU1 that removes sialic acids from glycoconjugates has been implicated in diverse cellular functions. Aberrant NEU1 activity is associated with various pathologies including lysosomal storage disorder sialidosis, autoimmune diseases and the malignancy and metastasis of cancer cells. We recently reported that NEU1 activity increases during 3T3-L1 adipogenesis and that it is higher in the epididymal fat of obese and diabetic mice. However, the precise functions of NEU1 in adipocytes have not been elucidated. Knockdown of NEU1 using siRNA transfection in 3T3-L1 adipocytes significantly decreased the mRNA expression and protein secretion of IL-6 and MCP-1 induced by LPS. The promoter activities of both IL-6 and MCP-1 as well as nuclear factor-kappa B (NF-κB) nuclear translocation were reduced in adipocytes transfected with an siRNA sequence that targets NEU1(siNEU1). NEU1 suppression using siNEU1 affected TLR4 sialylation. These findings suggest that NEU1 is involved in the production of IL-6 and MCP-1 in adipocytes possibly through TLR4/NF-κB signalling.


Assuntos
Adipócitos/metabolismo , NF-kappa B/metabolismo , Neuraminidase/metabolismo , Células 3T3-L1 , Animais , Células Cultivadas , Quimiocina CCL2/antagonistas & inibidores , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Perfilação da Expressão Gênica , Interleucina-6/antagonistas & inibidores , Interleucina-6/genética , Interleucina-6/metabolismo , Camundongos , Neuraminidase/deficiência , RNA Mensageiro/antagonistas & inibidores , RNA Mensageiro/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA