Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
1.
Infect Immun ; 89(11): e0025621, 2021 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-34424746

RESUMO

Clostridium perfringens type F strains causing nonfoodborne human gastrointestinal diseases (NFD) typically produce NanI sialidase as their major secreted sialidase. Type F NFDs can persist for several weeks, indicating their pathogenesis involves intestinal colonization, including vegetative cell growth and adherence, with subsequent sporulation that fosters enterotoxin production and release. We previously reported that NanI contributes to type F NFD strain adherence and growth using Caco-2 cells. However, Caco-2 cells make minimal amounts of mucus, which is significant because the intestines are coated with adherent mucus. Therefore, it was important to assess if NanI contributes to the growth and adherence of type F NFD strains in the presence of adherent mucus. Consequently, the current study first demonstrated greater growth of nanI-carrying versus non-nanI-carrying type F strains in the presence of HT29-MTX-E12 cells, which produce an adherent mucus layer, versus their parental HT29 cells, which make minimal mucus. Demonstrating the specific importance of NanI for this effect, type F NFD strain F4969 or a complementing strain grew and adhered better than an isogenic nanI null mutant in the presence of HT29-MTX-E12 cells versus HT29 cells. Those effects involved mucus production by HT29-MTX-E12 cells since mucus reduction using N-acetyl cysteine reduced F4969 growth and adherence. Consistent with those in vitro results, NanI contributed to growth of F4969 in the mouse small intestine. By demonstrating a growth and adherence role for NanI in the presence of adherent mucus, these results further support NanI as a potential virulence factor during type F NFDs.


Assuntos
Aderência Bacteriana/fisiologia , Clostridium perfringens/fisiologia , Intestinos/microbiologia , Muco/fisiologia , Neuraminidase/fisiologia , Células CACO-2 , Clostridium perfringens/crescimento & desenvolvimento , Células HT29 , Humanos , Fatores de Virulência/fisiologia
2.
Zhonghua Nan Ke Xue ; 26(8): 681-685, 2020 Aug.
Artigo em Chinês | MEDLINE | ID: mdl-33377727

RESUMO

OBJECTIVE: To investigate the effects of the enzyme activity of neuraminidase 1 (Neu1) on the biological behavior of prostate cancer PC3 and DU145 cell lines. METHODS: We detected the expression of Neul in the prostate cancer PC3 and DU145 cell lines by Western blot. Using sialidase inhibitors and antibody blocking, we suppressed the enzyme activity of Neu1 and then measured the proliferation and invasiveness of the two cell lines by CCK-8 and Transwell assay, respectively. RESULTS: No statistically significant difference was found in the Neu1 expression between the PC3 and DU145 cell lines. The proliferation and invasiveness of the two types of cells were both increased after inhibition of the Neu1 enzyme activity. CONCLUSIONS: The enzyme activity of Neu1 is correlated with the biological behavior of prostate cancer PC3 and DU145 cells and capable of inhibiting the proliferation and invasiveness of the two types of cells.


Assuntos
Proliferação de Células , Invasividade Neoplásica , Neuraminidase/fisiologia , Neoplasias da Próstata/enzimologia , Linhagem Celular Tumoral , Humanos , Masculino
3.
Cell Commun Signal ; 18(1): 44, 2020 03 12.
Artigo em Inglês | MEDLINE | ID: mdl-32164705

RESUMO

BACKGROUND: Sialic acids are widely distributed in animal tissues, and aberrantly expressed in a variety of cancer types. High expression of sialic acid contributes to tumor aggressiveness by promoting cell proliferation, migration, angiogenesis, and metastasis. Sialidases are responsible for removal of sialic acids from glycoproteins and glycolipids. METHODS: N-glycomics of bladder cancer cells were detected by MALDI-TOF mass spectrometry. Sialic acid modification in bladder cancer tissue was determined by lectin blot. The down-regulation of NEU1 in bladder cancer cells was determined by high resolution liquid chromatography mass spectrometry (HR LC-MS). The effects of sialidase NEU1 expression on proliferation and apoptosis of human bladder cancer cells were examined by western blot, RT-PCR, confocal imaging and flow cytometry. Moreover, the function of sialic acids on fibronectin-integrin α5ß1 interaction were assayed by immunoprecipitation and ELISA. The importance of NEU1 in tumor formation in vivo was performed using BALB/c-nu mice. Expression of NEU1 in primary human bladder cancer tissue samples was estimated using bladder cancer tissue microarray. RESULTS: (1) Downregulation of NEU1 was primarily responsible for aberrant expression of sialic acids in bladder cancer cells. (2) Decreased NEU1 expression was correlated with bladder cancer progression. (3) NEU1 overexpression enhanced apoptosis and reduced proliferation of bladder cancer cells. (4) NEU1 disrupted FN-integrin α5ß1 interaction and deactivated the Akt signaling pathway. (5) NEU1 significantly suppressed in vivo tumor formation in BALB/c-nu mice. CONCLUSIONS: Our data showed that NEU1 inhibited cancer cell proliferation, induced apoptosis, and suppressed tumor formation both in vitro and in vivo, by disrupting interaction of FN and integrin ß1 and inhibiting the Akt signaling pathway. Our observations indicate that NEU1 is an important modulator of the malignant properties of bladder cancer cells, and is a potential therapeutic target for prognosis and treatment of bladder cancer. Video Abstract.


Assuntos
Fibronectinas/metabolismo , Integrina alfa5beta1/metabolismo , Neuraminidase/fisiologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Neoplasias da Bexiga Urinária/metabolismo , Animais , Apoptose , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Progressão da Doença , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C
4.
J Chin Med Assoc ; 83(4): 337-344, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-31904658

RESUMO

Sialylation (the covalent addition of sialic acid to the terminal end of glycoproteins or glycans), tightly regulated cell- and microenvironment-specific process and orchestrated by sialyltransferases and sialidases (neuraminidases) family, is one of the posttranslational modifications, which plays an important biological role in the maintenance of normal physiology and involves many pathological dysfunctions. Glycans have roles in all the cancer hallmarks, referring to capabilities acquired during all steps of cancer development to initiate malignant transformation (a driver of a malignant genotype), enable cancer cells to survive, proliferate, and metastasize (a consequence of a malignant phenotype), which includes sustaining proliferative signaling, evading growth suppressor, resisting cell apoptosis, enabling replicative immortality, inducing angiogenesis, reprogramming of energy metabolism, evading tumor destruction, accumulating inflammatory microenvironment, and activating invasion and accelerating metastases. Regarding the important role of altered sialylation of cancers, further knowledge about the initiation and the consequences of altered sialylation pattern in tumor cells is needed, because all may offer a better chance for developing novel therapeutic strategy. In this review, we would like to update alteration of sialylation in ovarian cancers.


Assuntos
Neoplasias Ovarianas/metabolismo , Ácidos Siálicos/metabolismo , Biomarcadores Tumorais , Proteínas Sanguíneas/metabolismo , Feminino , Humanos , Neuraminidase/fisiologia , Antígeno Sialil Lewis X/análise , Sialiltransferases/fisiologia
5.
PLoS One ; 12(10): e0187289, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29088281

RESUMO

Adenocarcinoma of Non-Small Cell Lung Cancer (NSCLC) is a severe disease. Patients carrying EGFR mutations may benefit from EGFR targeted therapies (e.g.: gefitinib). Recently, it has been shown that sialidase NEU3 directly interacts and regulates EGFR. In this work, we investigate the effect of sialidase NEU3 overexpression on EGFR pathways activation and EGFR targeted therapies sensitivity, in a series of lung cancer cell lines. NEU3 overexpression, forced after transfection, does not affect NSCLC cell viability. We demonstrate that NEU3 overexpression stimulates the ERK pathway but this activation is completely abolished by gefitinib treatment. The Akt pathway is also hyper-activated upon NEU3 overexpression, but gefitinib is able only to decrease, and not to abolish, such activation. These findings indicate that NEU3 can act directly on the ERK pathway through EGFR and both directly and indirectly with respect to EGFR on the Akt pathway. Furthermore, we provide evidence that a healthy mucosa cell line (with EGFR wild-type gene sequence) is slightly sensitive to gefitinib, especially in the presence of NEU3 overexpression, thus hypothesizing that NEU3 overexpressing patients may benefit from EGFR targeted therapies also in absence of EGFR point mutations. Overall, the expression of NEU3 may be a novel diagnostic marker in NSCLC because, by its ability to stimulate EGFR downstream pathways with direct and indirect mechanisms, it may help in the identification of patients who can profit from EGFR targeted therapies in absence of EGFR activating mutations or from new combinations of EGFR and Akt inhibitors.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Receptores ErbB/fisiologia , Neuraminidase/fisiologia , Proteínas Tirosina Quinases/antagonistas & inibidores , Antineoplásicos/uso terapêutico , Western Blotting , Carcinoma Pulmonar de Células não Pequenas/fisiopatologia , Linhagem Celular Tumoral , Membrana Celular/enzimologia , Eletroforese em Gel de Poliacrilamida , Gefitinibe , Humanos , Neoplasias Pulmonares , Quinazolinas/uso terapêutico , Reação em Cadeia da Polimerase em Tempo Real , Transdução de Sinais/fisiologia
6.
BMC Microbiol ; 17(1): 178, 2017 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-28821225

RESUMO

BACKGROUND: Porphyromonas gingivalis is a major causative pathogen of chronic periodontitis. Within the inflammatory microenvironment, there exists extreme pH values, elevated temperatures and oxidative stress. Pathogens adapt to these stressful environmental conditions by regulating the transcription of virulence genes, modifying themselves with macromolecules and by aggregating and entering into a biofilm growth phase. Our previous study showed that the P. gingivalis sialidase can help cells obtain sialic acid from the environment, which is used to modify macromolecules on the surface of P. gingivalis cells. In this study, we compared the survival, virulence factors and biofilm formation of a sialidase-deficient strain (ΔPG0352) and the wild-type P. gingivalis W83 strain under various pH values, temperatures and oxidative stress conditions to identify the roles of sialidase in the adaptation of P. gingivalis to stressful conditions. RESULTS: Compared to the growth of the P. gingivalis W83 strain, the growth of the △PG0352 was more inhibited by oxidative stress (0.25 and 0.5 mM H2O2) and exhibited greater cell structure damage when treated with H2O2 as assessed by transmission electron microscopy. Both Lys-gingipain (Kgp) and Arg-gingipain (Rgp) activities were lower in the ΔPG0352 than those in the P. gingivalis W83 strain under all the assayed culture conditions. The lipopolysaccharide (LPS) activity of the W83 strain was higher than that of the ΔPG0352 under acidic conditions (pH 5.0), but no differences between the strains were observed under other conditions. Compared to the biofilms formed by P. gingivalis W83, those formed by the ΔPG0352 were decreased and discontinuous under acidic, alkaline and oxidative stress conditions. CONCLUSION: Compared to the P. gingivalis W83 strain, the survival, virulence and biofilm formation of the ΔPG0352 were decreased under stressful environmental conditions.


Assuntos
Biofilmes/crescimento & desenvolvimento , Neuraminidase/fisiologia , Porphyromonas gingivalis/fisiologia , Estresse Fisiológico , Virulência , Adaptação Biológica/fisiologia , Adesinas Bacterianas/metabolismo , Cisteína Endopeptidases/metabolismo , Genes Bacterianos , Cisteína Endopeptidases Gingipaínas , Peróxido de Hidrogênio/antagonistas & inibidores , Concentração de Íons de Hidrogênio , Lipopolissacarídeos/farmacologia , Viabilidade Microbiana/efeitos dos fármacos , Viabilidade Microbiana/genética , Microscopia Eletrônica de Transmissão , Mutação , Neuraminidase/genética , Estresse Oxidativo , Porphyromonas gingivalis/efeitos dos fármacos , Porphyromonas gingivalis/genética , Temperatura , Virulência/genética , Fatores de Virulência/genética
7.
Infect Immun ; 84(10): 2922-32, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27481242

RESUMO

Streptococcus pneumoniae is an opportunistic pathogen that colonizes the nasopharynx. Herein we show that carbon availability is distinct between the nasopharynx and bloodstream of adult humans: glucose is absent from the nasopharynx, whereas galactose is abundant. We demonstrate that pneumococcal neuraminidase A (NanA), which cleaves terminal sialic acid residues from host glycoproteins, exposed galactose on the surface of septal epithelial cells, thereby increasing its availability during colonization. We observed that S. pneumoniae mutants deficient in NanA and ß-galactosidase A (BgaA) failed to form biofilms in vivo despite normal biofilm-forming abilities in vitro Subsequently, we observed that glucose, sucrose, and fructose were inhibitory for biofilm formation, whereas galactose, lactose, and low concentrations of sialic acid were permissive. Together these findings suggested that the genes involved in biofilm formation were under some form of carbon catabolite repression (CCR), a regulatory network in which genes involved in the uptake and metabolism of less-preferred sugars are silenced during growth with preferred sugars. Supporting this notion, we observed that a mutant deficient in pyruvate oxidase, which converts pyruvate to acetyl-phosphate under non-CCR-inducing growth conditions, was unable to form biofilms. Subsequent comparative transcriptome sequencing (RNA-seq) analyses of planktonic and biofilm-grown pneumococci showed that metabolic pathways involving the conversion of pyruvate to acetyl-phosphate and subsequently leading to fatty acid biosynthesis were consistently upregulated during diverse biofilm growth conditions. We conclude that carbon availability in the nasopharynx impacts pneumococcal biofilm formation in vivo Additionally, biofilm formation involves metabolic pathways not previously appreciated to play an important role.


Assuntos
Biofilmes/crescimento & desenvolvimento , Metabolismo dos Carboidratos/fisiologia , Carboidratos/farmacologia , Galactose/farmacocinética , Neuraminidase/fisiologia , Infecções Pneumocócicas/microbiologia , Streptococcus pneumoniae/fisiologia , Análise de Variância , Animais , Biofilmes/efeitos dos fármacos , Modelos Animais de Doenças , Células Epiteliais/metabolismo , Feminino , Galactose/metabolismo , Galactose/farmacologia , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Ácido N-Acetilneuramínico/metabolismo , Líquido da Lavagem Nasal/química , Septo Nasal/metabolismo , Septo Nasal/microbiologia , Nasofaringe/metabolismo , Nasofaringe/microbiologia , Neuraminidase/metabolismo , Infecções Pneumocócicas/metabolismo , Streptococcus pneumoniae/efeitos dos fármacos , beta-Galactosidase/deficiência , beta-Galactosidase/metabolismo
8.
Yakugaku Zasshi ; 135(12): 1341-8, 2015.
Artigo em Japonês | MEDLINE | ID: mdl-26632149

RESUMO

Sialidase removes sialic acid residues from sialoglycoconjugates such as glycoproteins and glycolipids. Since sialic acid plays crucial roles in synaptic plasticity and memory in the hippocampus, the regulation of sialyl signaling by sialidase is also necessary for neural functions. However, since mammalian sialidase activity is remarkably weak, it has been difficult to detect sialidase activity in mammalian tissues. Determination of the distribution of sialidase activity in living mammalian tissues would provide much valuable information for understanding the roles of sialidase in physiological functions. Therefore, we synthesized a novel benzothiazolylphenol-based sialic acid derivative (BTP-Neu5Ac) as a fluorescent sialidase substrate. After cleavage of BTP-Neu5Ac, which is water soluble and shows little fluorescence, with sialidase, the water-insoluble fluorophore benzothiazolylphenol (BTP) released from BTP-Neu5Ac stains tissue and shows bright fluorescence. BTP-Neu5Ac can visualize sialidase activity in brain tissue with high levels of sensitivity and specificity. The sialidase expression level is markedly high in various human cancers such as colon, renal, prostate, and ovarian cancers. BTP-Neu5Ac can detect human colon cancers sensitively. Thus, BTP-Neu5Ac is useful not only for physiological research but also as a cancer probe. BTP-Neu5Ac is now being used in virology research. In this review, methods for histochemical imaging of sialidase activity and the role of sialidase in hippocampal memory are described based on the author's study of multidimensional analysis of hippocampal excitatory neurotransmission and development of analytical tools for glycans, which was awarded a prize by the Tokai branch of the Pharmaceutical Society of Japan.


Assuntos
Hipocampo/fisiologia , Neuraminidase/análise , Polissacarídeos/análise , Transmissão Sináptica/fisiologia , Animais , Neoplasias do Colo , Humanos , Memória/fisiologia , Ácido N-Acetilneuramínico , Neuraminidase/fisiologia
9.
Biochem J ; 470(1): 131-44, 2015 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-26251452

RESUMO

Gangliosides are sialic acid-containing glycosphingolipids mainly expressed at the outer leaflet of the plasma membrane. Sialidase NEU3 is a key enzyme in the catabolism of gangliosides with its up-regulation having been observed in human cancer cells. In the case of CME (clathrin-mediated endocytosis), although this has been widely studied, the role of NEU3 and gangliosides in this cellular process has not yet been established. In the present study, we found an increased internalization of Tf (transferrin), the archetypical cargo for CME, in cells expressing complex gangliosides with high levels of sialylation. The ectopic expression of NEU3 led to a drastic decrease in Tf endocytosis, suggesting the participation of gangliosides in this process. However, the reduction in Tf endocytosis caused by NEU3 was still observed in glycosphingolipid-depleted cells, indicating that NEU3 could operate in a way that is independent of its action on gangliosides. Additionally, internalization of α2-macroglobulin and low-density lipoprotein, other typical ligands in CME, was also decreased in NEU3-expressing cells. In contrast, internalization of cholera toxin ß-subunit, which is endocytosed by both clathrin-dependent and clathrin-independent mechanisms, remained unaltered. Kinetic assays revealed that NEU3 caused a reduction in the sorting of endocytosed Tf to early and recycling endosomes, with the Tf binding at the cell surface being also reduced. NEU3-expressing cells showed an altered subcellular distribution of clathrin adaptor AP-2 (adaptor protein 2), but did not reveal any changes in the membrane distribution of clathrin, PtdIns(4,5)P2 or caveolin-1. Overall, these results suggest a specific and novel role of NEU3 in CME.


Assuntos
Membrana Celular/metabolismo , Clatrina/metabolismo , Endocitose/fisiologia , Neuraminidase/fisiologia , Animais , Células CHO , Células COS , Galinhas , Chlorocebus aethiops , Cricetinae , Cricetulus , Humanos , Ligação Proteica/fisiologia
10.
Nat Commun ; 4: 2734, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24225533

RESUMO

Alzheimer's disease (AD) belongs to a category of adult neurodegenerative conditions, which are associated with intracellular and extracellular accumulation of neurotoxic protein aggregates. Understanding how these aggregates are formed, secreted and propagated by neurons has been the subject of intensive research, but so far no preventive or curative therapy for AD is available, and clinical trials have been largely unsuccessful. Here we show that deficiency of the lysosomal sialidase NEU1 leads to the spontaneous occurrence of an AD-like amyloidogenic process in mice. This involves two consecutive events linked to NEU1 loss-of-function--accumulation and amyloidogenic processing of an oversialylated amyloid precursor protein in lysosomes, and extracellular release of Aß peptides by excessive lysosomal exocytosis. Furthermore, cerebral injection of NEU1 in an established AD mouse model substantially reduces ß-amyloid plaques. Our findings identify an additional pathway for the secretion of Aß and define NEU1 as a potential therapeutic molecule for AD.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Exocitose/fisiologia , Lisossomos/metabolismo , Mucolipidoses/genética , Neuraminidase/genética , Animais , Encéfalo/embriologia , Calcimicina/metabolismo , Linhagem Celular , Dependovirus/metabolismo , Hipocampo/embriologia , Hipocampo/metabolismo , Hipocampo/ultraestrutura , Humanos , Camundongos , Camundongos Transgênicos , Neuraminidase/fisiologia , Neurônios/metabolismo , Fatores de Risco
11.
Cell Microbiol ; 15(8): 1357-66, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23414299

RESUMO

Chronic Chagas cardiomyopathy (CCC), caused by the obligate intracellular protozoan parasite Trypanosoma cruzi, is a major cause of morbidity and mortality in Latin America. CCC begins when T. cruzi enters cardiac cells for intracellular multiplication and differentiation, a process that starts with recognition of host-cell entry receptors. However, the nature of these surface molecules and corresponding parasite counter-receptor(s) is poorly understood. Here we show that antibodies against neurotrophin (NT) receptor TrkC, but not against family members TrkA and TrkB, prevent T. cruzi from invading primary cultures of cardiomyocytes and cardiac fibroblasts. Invasion is also selectively blocked by the TrkC ligand NT-3, and by antagonists of Trk autophosphorylation and downstream signalling. Therefore, these results indicate that T. cruzi gets inside cardiomyocytes and cardiac fibroblasts by activating TrkC preferentially over TrkA. Accordingly, short hairpin RNA interference of TrkC (shTrkC), but not TrkA, selectively prevents T. cruzi from entering cardiac cells. Additionally, T. cruzi parasite-derived neurotrophic factor (PDNF)/trans-sialidase, a TrkC-binding protein, but not family member gp85, blocks entry dose-dependently, underscoring the specificity of PDNF as TrkC counter-receptor in cardiac cell invasion. In contrast to invasion, competitive and shRNA inhibition studies demonstrate that T. cruzi-PDNF recognition of TrkA, but not TrkC on primary cardiomyocytes and the cardiomyocyte cell line H9c2 protects the cells against oxidative stress. Thus, this study shows that T. cruzi via PDNF favours neurotrophin receptor TrkC for cardiac cell entry and TrkA for cardiomyocyte protection against oxidative stress, and suggests a new therapeutic opportunity in PDNF and/or fragments thereof for CCC therapy as entry inhibitors and/or cardioprotection agonists.


Assuntos
Fibroblastos/parasitologia , Miócitos Cardíacos/parasitologia , Estresse Oxidativo/fisiologia , Receptor trkA/fisiologia , Receptor trkC/fisiologia , Trypanosoma cruzi/patogenicidade , Animais , Células Cultivadas , Cardiomiopatia Chagásica , Modelos Animais de Doenças , Fibroblastos/patologia , Fibroblastos/fisiologia , Glicoproteínas/fisiologia , Interações Hospedeiro-Parasita/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Miócitos Cardíacos/patologia , Miócitos Cardíacos/fisiologia , Neuraminidase/fisiologia , RNA Interferente Pequeno/farmacologia , Receptor trkC/antagonistas & inibidores , Receptor trkC/efeitos dos fármacos , Trypanosoma cruzi/fisiologia
12.
Mol Oral Microbiol ; 27(4): 270-83, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22759312

RESUMO

Streptococcus pneumoniae colonizes the upper respiratory tract from where the organisms may disseminate systemically to cause life threatening infections. The mechanisms by which pneumococci colonize epithelia are not understood, but neuraminidase A (NanA) has a major role in promoting growth and survival in the upper respiratory tract. In this article we show that mutants of S. pneumoniae D39 deficient in NanA or neuraminidase B (NanB) are abrogated in adherence to three epithelial cell lines, and to primary nasopharyngeal cells. Adherence levels were partly restored by nanA complementation in trans. Enzymic activity of NanA was shown to be necessary for pneumococcal adherence to epithelial cells, and adherence of the nanA mutant was restored to wild-type level by pre-incubation of epithelial cells with Lactococcus lactis cells expressing NanA. Pneumococcal nanA or nanB mutants were deficient in biofilm formation, while expression of NanA on L. lactis or Streptococcus gordonii promoted biofilm formation by these heterologous host organisms. The results suggest that NanA is an enzymic factor mediating adherence to epithelial cells by decrypting receptors for adhesion, and functions at least in part as an adhesin in biofilm formation. Neuraminidase A thus appears to play multiple temporal roles in pneumococcal infection, from adherence to host tissues, colonization, and community development, to systemic spread and crossing of the blood-brain barrier.


Assuntos
Proteínas de Bactérias/fisiologia , Neuraminidase/fisiologia , Infecções Pneumocócicas/enzimologia , Streptococcus pneumoniae/enzimologia , Fatores de Virulência , Aderência Bacteriana/fisiologia , Proteínas de Bactérias/genética , Biofilmes/crescimento & desenvolvimento , Células Cultivadas , Células Epiteliais/microbiologia , Humanos , Mutação , Nasofaringe/citologia , Neuraminidase/genética , Ligação Proteica , Receptores de Superfície Celular/metabolismo , Sistema Respiratório/citologia , Streptococcus pneumoniae/genética
13.
Glycobiology ; 22(7): 880-96, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22377912

RESUMO

Sialic acids are terminal acidic monosaccharides, which influence the chemical and biological features of glycoconjugates. Their removal catalyzed by a sialidase modulates various biological processes through change in conformation and creation or loss of binding sites of functional molecules. Sialidases exist widely in vertebrates and also in a variety of microorganisms. Recent research on mammalian sialidases has provided evidence for great importance of these enzymes in various cellular functions, including lysosomal catabolism, whereas microbial sialidases appear to play roles limited to nutrition and pathogenesis. Four types of mammalian sialidases have been identified and characterized to date, designated as NEU1, NEU2, NEU3 and NEU4. They are encoded by different genes and differ in major subcellular localization and enzymatic properties including substrate specificity, and each has been found to play a unique role depending on its particular properties. This review is an attempt to concisely summarize current knowledge concerning mammalian sialidases, with a special focus on their properties and physiological and pathological roles in cellular functions.


Assuntos
Neuraminidase/fisiologia , Sequência de Aminoácidos , Animais , Sequência Conservada , Componentes do Gene , Humanos , Dados de Sequência Molecular , Neoplasias/enzimologia , Neuraminidase/genética , Neuraminidase/metabolismo , Neuritos/enzimologia , Regeneração , Transdução de Sinais , Especificidade por Substrato
14.
Blood ; 119(5): 1263-73, 2012 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-22101895

RESUMO

When refrigerated platelets are rewarmed, they secrete active sialidases, including the lysosomal sialidase Neu1, and express surface Neu3 that remove sialic acid from platelet von Willebrand factor receptor (VWFR), specifically the GPIbα subunit. The recovery and circulation of refrigerated platelets is greatly improved by storage in the presence of inhibitors of sialidases. Desialylated VWFR is also a target for metalloproteinases (MPs), because GPIbα and GPV are cleaved from the surface of refrigerated platelets. Receptor shedding is inhibited by the MP inhibitor GM6001 and does not occur in Adam17(ΔZn/ΔZn) platelets expressing inactive ADAM17. Critically, desialylation in the absence of MP-mediated receptor shedding is sufficient to cause the rapid clearance of platelets from circulation. Desialylation of platelet VWFR therefore triggers platelet clearance and primes GPIbα and GPV for MP-dependent cleavage.


Assuntos
Plaquetas/fisiologia , Glicoproteínas de Membrana/metabolismo , Metaloproteases/metabolismo , Neuraminidase/metabolismo , Refrigeração , Proteínas ADAM/metabolismo , Proteínas ADAM/fisiologia , Proteína ADAM17 , Animais , Plaquetas/metabolismo , Preservação de Sangue/métodos , Ativação Enzimática , Glicosilação , Humanos , Masculino , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/fisiologia , Metaloproteases/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Neuraminidase/fisiologia , Complexo Glicoproteico GPIb-IX de Plaquetas , Processamento de Proteína Pós-Traducional/fisiologia , Proteólise , Refrigeração/métodos , Fator de von Willebrand/metabolismo
15.
Microbiol Immunol ; 54(10): 584-95, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21118296

RESUMO

A sialidase gene of Streptococcus intermedius was cloned. It was most similar to nanA, a major sialidase gene in Streptococcus pneumoniae, and was expressed in Escherichia coli. Since the gene-knockout S. intermedius strain lost detectable sialidase activity, the gene might code, either solely or mainly, the glycosidase in the bacterial genome. Polymerase chain reaction using the primers for the nanA homologue in S. intermedius (described as nanA below) showed that this sialidase gene was commonly distributed within the isolates of S. intermedius, but not found in the strains of other species among the anginosus group. In biofilm formation assay under cultivation with mucin, the nanA-deleted S. intermedius maintained the amount of biofilm for 72 hr, while that of the parent strain decreased during incubation from 24 to 72 hr. Since sialidase activity in the parent strain increased during that time period, sialidase might contribute to the degradation of biofilm under sialic acid-rich conditions. When S. intermedius was added into the HepG2 hepatoma culture, the calculated disassociation constant (K(d)) of EDTA-releasable bacterial adhesion to the cells was higher in the nanA-deleted strain than in the parent. Furthermore, the rate constant, assuming endocytosis of the bacterium mediated by ASGP-R in HepG2 cells, seemed to be increased by sialidase pretreatment of the bacterial cells before addition to the cell culture. According to the results, modification of sugar chains by sialidase on the bacterial surface and in the surrounding environment might influence both bacterial interaction and host-bacterial interaction in S. intermedius.


Assuntos
Neuraminidase/fisiologia , Streptococcus intermedius/enzimologia , Streptococcus intermedius/patogenicidade , Fatores de Virulência/fisiologia , Biofilmes , Células Hep G2 , Humanos , Neuraminidase/genética
16.
PLoS Pathog ; 6(10): e1001136, 2010 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-20949074

RESUMO

Transforming growth factor-beta (TGF-ß), a multifunctional cytokine regulating several immunologic processes, is expressed by virtually all cells as a biologically inactive molecule termed latent TGF-ß (LTGF-ß). We have previously shown that TGF-ß activity increases during influenza virus infection in mice and suggested that the neuraminidase (NA) protein mediates this activation. In the current study, we determined the mechanism of activation of LTGF-ß by NA from the influenza virus A/Gray Teal/Australia/2/1979 by mobility shift and enzyme inhibition assays. We also investigated whether exogenous TGF-ß administered via a replication-deficient adenovirus vector provides protection from H5N1 influenza pathogenesis and whether depletion of TGF-ß during virus infection increases morbidity in mice. We found that both the influenza and bacterial NA activate LTGF-ß by removing sialic acid motifs from LTGF-ß, each NA being specific for the sialic acid linkages cleaved. Further, NA likely activates LTGF-ß primarily via its enzymatic activity, but proteases might also play a role in this process. Several influenza A virus subtypes (H1N1, H1N2, H3N2, H5N9, H6N1, and H7N3) except the highly pathogenic H5N1 strains activated LTGF-ß in vitro and in vivo. Addition of exogenous TGF-ß to H5N1 influenza virus-infected mice delayed mortality and reduced viral titers whereas neutralization of TGF-ß during H5N1 and pandemic 2009 H1N1 infection increased morbidity. Together, these data show that microbe-associated NAs can directly activate LTGF-ß and that TGF-ß plays a pivotal role protecting the host from influenza pathogenesis.


Assuntos
Virus da Influenza A Subtipo H5N1/patogenicidade , Influenza Humana/metabolismo , Neuraminidase/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Células Cultivadas , Embrião de Galinha , Cães , Ativação Enzimática/fisiologia , Humanos , Virus da Influenza A Subtipo H5N1/fisiologia , Influenza Humana/virologia , Camundongos , Camundongos Endogâmicos BALB C , Neuraminidase/isolamento & purificação , Neuraminidase/farmacologia , Neuraminidase/fisiologia , Infecções por Orthomyxoviridae/metabolismo , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia , Fator de Crescimento Transformador beta/fisiologia
17.
Infect Immun ; 77(10): 4421-8, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19651873

RESUMO

The essential toxin in Clostridium perfringens-mediated gas gangrene or clostridial myonecrosis is alpha-toxin, although other toxins and extracellular enzymes may also be involved. In many bacterial pathogens extracellular sialidases are important virulence factors, and it has been suggested that sialidases may play a role in gas gangrene. C. perfringens strains have combinations of three different sialidase genes, two of which, nanI and nanJ, encode secreted sialidases. The nanI and nanJ genes were insertionally inactivated by homologous recombination in derivatives of sequenced strain 13 and were shown to encode two functional secreted sialidases, NanI and NanJ. Analysis of these derivatives showed that NanI was the major sialidase in this organism. Mutation of nanI resulted in loss of most of the secreted sialidase activity, and the residual activity was eliminated by subsequent mutation of the nanJ gene. Only a slight reduction in the total sialidase activity was observed in a nanJ mutant. Cytotoxicity assays using the B16 melanoma cell line showed that supernatants containing NanI or overexpressing NanJ enhanced alpha-toxin-mediated cytotoxicity. Finally, the ability of nanI, nanJ, and nanIJ mutants to cause disease was assessed in a mouse myonecrosis model. No attenuation of virulence was observed for any of these strains, providing evidence that neither the NanI sialidase nor the NanJ sialidase is essential for virulence.


Assuntos
Proteínas de Bactérias/fisiologia , Clostridium perfringens/enzimologia , Clostridium perfringens/patogenicidade , Gangrena Gasosa/microbiologia , Neuraminidase/fisiologia , Fatores de Virulência/fisiologia , Animais , Proteínas de Bactérias/genética , Linhagem Celular Tumoral , Sobrevivência Celular , Técnicas de Inativação de Genes , Camundongos , Camundongos Endogâmicos BALB C , Mutagênese Insercional , Neuraminidase/genética , Análise de Sobrevida , Virulência , Fatores de Virulência/genética
18.
Infect Immun ; 77(9): 3588-95, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19528219

RESUMO

The underlying mechanisms of the epidemiological association between influenza virus infections and Neisseria meningitidis invasive infections are not fully understood. Here we report that adhesion of N. meningitidis to human Hec-1-B epithelial cells is enhanced by influenza A virus (IAV) infection. A potential role of the viral neuraminidase (NA) in facilitating meningococcal adhesion to influenza virus-infected epithelial cells was examined. Expression of a recombinant IAV NA in Hec-1-B human epithelial cells increased the adhesion of strains of N. meningitidis belonging to the sialic acid-containing capsular serogroups B, C, and W135 but not to the mannosamine phosphate-containing capsular serogroup A. Adhesion enhancement was not observed with an inactive NA mutant or in the presence of an NA inhibitor (zanamivir). Furthermore, purified IAV NA was shown to cleave sialic acid-containing capsular polysaccharides of N. meningitidis. On the whole, our findings suggest that a direct interaction between the NA of IAV and the capsule of N. meningitidis enhances bacterial adhesion to cultured epithelial cells, most likely through cleavage of capsular sialic acid-containing polysaccharides. A better understanding of the association between IAV and invasive meningococcal infections should help to set up improved control strategies against these seasonal dual viral-bacterial infections.


Assuntos
Aderência Bacteriana , Cápsulas Bacterianas/fisiologia , Vírus da Influenza A Subtipo H1N1/fisiologia , Vírus da Influenza A Subtipo H3N2/fisiologia , Neisseria meningitidis/fisiologia , Neuraminidase/fisiologia , Ácidos Siálicos/fisiologia , Linhagem Celular Tumoral , Células Epiteliais/microbiologia , Humanos , Neuraminidase/genética
19.
J Biochem ; 144(3): 279-85, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18632803

RESUMO

Mammalian sialidases, glycosidases responsible for the removal of sialic acids from glycoproteins and glycolipids, has been implicated to participate in many biological processes as well as in lysosomal catabolism. Among those forms identified to date, plasma membrane-associated sialidase, Neu3, is a key enzyme in degradation of gangliosides, for which it exhibits a special substrate preference. This sialidase has been shown to control transmembrane signalling for many cellular processes, including cell differentiation, cell growth and apoptosis, and human orthologue NEU3 is markedly up-regulated in various cancers. It is known to suppress apoptosis in cancer cells. Furthermore, its overexpression causes impaired glucose tolerance and hyper-insulinaemia together with overproduction of insulin in enlarged islets in the transgenic mice. The present review primarily summarizes our recent results, focusing on Neu3 as a regulator of transmembrane signalling.


Assuntos
Membrana Celular/enzimologia , Regulação Enzimológica da Expressão Gênica , Neuraminidase/biossíntese , Neuraminidase/química , Animais , Apoptose , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Humanos , Camundongos , Camundongos Transgênicos , Modelos Biológicos , Neoplasias/metabolismo , Neuraminidase/metabolismo , Neuraminidase/fisiologia , Neuritos/metabolismo , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA