Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 199
Filtrar
Mais filtros

País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
PLoS Pathog ; 17(7): e1009381, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34197564

RESUMO

Clearance of viral infections, such as SARS-CoV-2 and influenza A virus (IAV), must be fine-tuned to eliminate the pathogen without causing immunopathology. As such, an aggressive initial innate immune response favors the host in contrast to a detrimental prolonged inflammation. The complement pathway bridges innate and adaptive immune system and contributes to the response by directly clearing pathogens or infected cells, as well as recruiting proinflammatory immune cells and regulating inflammation. However, the impact of modulating complement activation in viral infections is still unclear. In this work, we targeted the complement decay-accelerating factor (DAF/CD55), a surface protein that protects cells from non-specific complement attack, and analyzed its role in IAV infections. We found that DAF modulates IAV infection in vivo, via an interplay with the antigenic viral proteins hemagglutinin (HA) and neuraminidase (NA), in a strain specific manner. Our results reveal that, contrary to what could be expected, DAF potentiates complement activation, increasing the recruitment of neutrophils, monocytes and T cells. We also show that viral NA acts on the heavily sialylated DAF and propose that the NA-dependent DAF removal of sialic acids exacerbates complement activation, leading to lung immunopathology. Remarkably, this mechanism has no impact on viral loads, but rather on the host resilience to infection, and may have direct implications in zoonotic influenza transmissions.


Assuntos
Antígenos CD55/fisiologia , Vírus da Influenza A Subtipo H1N1/isolamento & purificação , Pulmão/imunologia , Viremia/imunologia , Animais , Líquido da Lavagem Broncoalveolar/imunologia , Antígenos CD55/química , Antígenos CD55/deficiência , Quimiotaxia de Leucócito , Ativação do Complemento , Glicoproteínas de Hemaglutininação de Vírus da Influenza/fisiologia , Adaptação ao Hospedeiro , Especificidade de Hospedeiro , Interações Hospedeiro-Patógeno , Vírus da Influenza A Subtipo H1N1/enzimologia , Vírus da Influenza A Subtipo H1N1/patogenicidade , Vírus da Influenza A Subtipo H1N1/fisiologia , Interferon gama/análise , Pulmão/patologia , Pulmão/virologia , Camundongos , Camundongos Endogâmicos C57BL , Ácido N-Acetilneuramínico , Neuraminidase/fisiologia , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/patologia , Carga Viral , Proteínas Virais/fisiologia , Virulência , Replicação Viral , Redução de Peso
2.
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
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.
Zhonghua Nan Ke Xue ; 26(8): 681-685, 2020 Aug.
Artigo em Zh | 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
5.
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
6.
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
7.
PLoS Pathog ; 10(9): e1004366, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25211026

RESUMO

The central event underlying prion diseases involves conformational change of the cellular form of the prion protein (PrP(C)) into the disease-associated, transmissible form (PrP(Sc)). Pr(PC) is a sialoglycoprotein that contains two conserved N-glycosylation sites. Among the key parameters that control prion replication identified over the years are amino acid sequence of host PrP(C) and the strain-specific structure of PrPSc. The current work highlights the previously unappreciated role of sialylation of PrP(C) glycans in prion pathogenesis, including its role in controlling prion replication rate, infectivity, cross-species barrier and PrP(Sc) glycoform ratio. The current study demonstrates that undersialylated PrP(C) is selected during prion amplification in Protein Misfolding Cyclic Amplification (PMCAb) at the expense of oversialylated PrP(C). As a result, PMCAb-derived PrP(Sc) was less sialylated than brain-derived PrP(Sc). A decrease in PrPSc sialylation correlated with a drop in infectivity of PMCAb-derived material. Nevertheless, enzymatic de-sialylation of PrP(C) using sialidase was found to increase the rate of PrP(Sc) amplification in PMCAb from 10- to 10,000-fold in a strain-dependent manner. Moreover, de-sialylation of PrP(C) reduced or eliminated a species barrier of for prion amplification in PMCAb. These results suggest that the negative charge of sialic acid controls the energy barrier of homologous and heterologous prion replication. Surprisingly, the sialylation status of PrP(C) was also found to control PrP(Sc) glycoform ratio. A decrease in Pr(PC) sialylation levels resulted in a higher percentage of the diglycosylated glycoform in PrP(Sc). 2D analysis of charge distribution revealed that the sialylation status of brain-derived PrP(C) differed from that of spleen-derived PrP(C). Knocking out lysosomal sialidase Neu1 did not change the sialylation status of brain-derived PrP(C), suggesting that Neu1 is not responsible for desialylation of PrP(C). The current work highlights previously unappreciated role of PrP(C) sialylation in prion diseases and opens multiple new research directions, including development of new therapeutic approaches.


Assuntos
Ácido N-Acetilneuramínico/metabolismo , Proteínas PrPC/metabolismo , Proteínas PrPSc/metabolismo , Doenças Priônicas/transmissão , Dobramento de Proteína , Animais , Western Blotting , Encéfalo/metabolismo , Encéfalo/patologia , Glicosilação , Masculino , Mesocricetus , Camundongos , Camundongos Knockout , Neuraminidase/metabolismo , Neuraminidase/fisiologia , Proteínas PrPC/química , Proteínas PrPC/patogenicidade , Proteínas PrPSc/química , Proteínas PrPSc/patogenicidade , Doenças Priônicas/metabolismo , Doenças Priônicas/patologia , Baço/metabolismo , Baço/patologia
8.
Curr Top Microbiol Immunol ; 386: 275-99, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25033754

RESUMO

Neuraminidase (NA) is the second most abundant influenza surface glycoprotein and contributes to virus replication in several ways, most notably by removing sialic acids from the host and viral glycoproteins, releasing newly formed virus particles from infected cells. Antibodies that block this enzyme activity restrict virus replication in vitro. This chapter describes foundational epidemiologic and human influenza challenge studies that provide evidence of an association between NA inhibiting antibodies and resistance to disease. Mouse challenge studies show that while NA immunity is infection-permissive, NA-specific antibodies attenuate infection and prevent severe disease. NA immunity is most effective against homologous viruses but there is substantial protection against viruses with a heterologous NA (different lineage within a NA subtype). Monoclonal antibodies specific for conserved antigenic domains of subtype N1 protect against seasonal and pandemic H1N1 as well as H5N1 virus challenge. Clinical studies demonstrate that licensed seasonal vaccines contain immunogenic amounts of NA, but the contribution of this immunity to vaccine efficacy is currently not known. New types of influenza vaccines could be designed to elicit NA immunity. Because NA induces heterologous immunity, it could be an important constituent of universal influenza vaccines that aim to protect against unexpected emerging viruses.


Assuntos
Vacinas contra Influenza/imunologia , Neuraminidase/imunologia , Orthomyxoviridae/enzimologia , Animais , Humanos , Camundongos , Neuraminidase/química , Neuraminidase/fisiologia
9.
Vet Res ; 47(1): 53, 2016 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-27160077

RESUMO

Canine influenza virus (CIV) is a newly identified, highly contagious respiratory pathogen in dogs. Recent studies indicate that avian-origin H3N2 CIV are circulating in Chinese dogs. To investigate the effects of a two-amino acid (2-aa) insertion naturally occurring at the distal end of the neuraminidase (NA) stalk found in Chinese isolates since 2010 on virus replication and virulence, we rescued the CIV strain, A/canine/Jiangsu/06/2011(H3N2) and its NA mutant without the 2-aa insertion using reverse genetics. The NA stalk length affected virus growth in cell culture. Compared to the short stalk strain (without 2-aa insertion), the long stalk strain (with 2-aa insertion) exhibited higher peak titers and greater yields in Madin-Darby canine kidney (MDCK) cells, chicken embryo fibroblasts and canine bronchiolar epithelial cells, as well as much larger plaques in MDCK cell monolayers. Furthermore, mice inoculated with the long stalk strain showed more severe pathologic damage in lung and higher proportion of detectable viral RNA in tissues. The long stalk strain induced local IFN-γ production with faster kinetics and higher levels in mice. However, in chickens, the two viral strains showed no significant difference with nearly the same proportion of detectable viral RNA loads in tissues. These observations suggest that the 2-aa insertion in the NA stalk acquired by avian-origin H3N2 CIV helps to enhance viral replication and is likely a result of adaptive evolution in canine hosts.


Assuntos
Doenças do Cão/virologia , Vírus da Influenza A Subtipo H3N2/fisiologia , Neuraminidase/fisiologia , Replicação Viral/fisiologia , Animais , Células Cultivadas , Embrião de Galinha/virologia , Galinhas/virologia , Cães , Feminino , Citometria de Fluxo , Imunofluorescência , Vírus da Influenza A Subtipo H3N2/genética , Camundongos , Camundongos Endogâmicos BALB C , Mutagênese Insercional/genética , Mutagênese Insercional/fisiologia , Mutagênese Sítio-Dirigida , Neuraminidase/genética , Infecções por Orthomyxoviridae/veterinária , Infecções por Orthomyxoviridae/virologia , Replicação Viral/genética
10.
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
11.
Neural Plast ; 2015: 908190, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25802763

RESUMO

Neuraminidase (NEU) is a key enzyme that cleaves negatively charged sialic acid residues from membrane proteins and lipids. Clinical and basic science studies have shown that an imbalance in NEU metabolism or changes in NEU activity due to various pathological conditions parallel with behavior and cognitive impairment. It has been suggested that the decreases of NEU activity could cause serious neurological consequences. However, there is a lack of direct evidences that modulation of endogenous NEU activity can impair neuronal function. Using combined rat entorhinal cortex/hippocampal slices and a specific inhibitor of NEU, 2-deoxy-2,3-dehydro-N-acetylneuraminic acid (NADNA), we examined the effect of downregulation of NEU activity on different forms of synaptic plasticity in the hippocampal CA3-to-CA1 network. We show that NEU inhibition results in a significant decrease in long-term potentiation (LTP) and an increase in short-term depression. Synaptic depotentiation restores LTP in NADNA-pretreated slices to the control level. These data suggest that short-term NEU inhibition produces the LTP-like effect on neuronal network, which results in damping of further LTP induction. Our findings demonstrate that downregulation of NEU activity could have a major impact on synaptic plasticity and provide a new insight into the cellular mechanism underlying behavioral and cognitive impairment associated with abnormal metabolism of NEU.


Assuntos
Hipocampo/enzimologia , Hipocampo/fisiologia , Neuraminidase/fisiologia , Plasticidade Neuronal , Transmissão Sináptica , Animais , Hipocampo/efeitos dos fármacos , Neuraminidase/antagonistas & inibidores , Plasticidade Neuronal/efeitos dos fármacos , Ratos , Ratos Wistar , Transmissão Sináptica/efeitos dos fármacos
12.
J Gen Virol ; 95(Pt 6): 1193-1210, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24584475

RESUMO

Typical avian influenza A viruses are restricted from replicating efficiently and causing disease in humans. However, an avian virus can become adapted to humans by mutating or recombining with currently circulating human viruses. These viruses have the potential to cause pandemics in an immunologically naïve human population. It is critical that we understand the molecular basis of host-range restriction and how this can be overcome. Here, we review our current understanding of the mechanisms by which influenza viruses adapt to replicate efficiently in a new host. We predominantly focus on the influenza polymerase, which remains one of the least understood host-range barriers.


Assuntos
Vírus da Influenza A/patogenicidade , Adaptação Fisiológica , Animais , Aves , RNA Polimerases Dirigidas por DNA/genética , RNA Polimerases Dirigidas por DNA/fisiologia , Genes Virais , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Glicoproteínas de Hemaglutininação de Vírus da Influenza/fisiologia , Especificidade de Hospedeiro/genética , Especificidade de Hospedeiro/imunologia , Especificidade de Hospedeiro/fisiologia , Humanos , Imunidade Inata , Vírus da Influenza A/genética , Vírus da Influenza A/fisiologia , Influenza Aviária/imunologia , Influenza Aviária/virologia , Influenza Humana/imunologia , Influenza Humana/virologia , Mutação , Neuraminidase/genética , Neuraminidase/fisiologia , Nucleoproteínas/genética , Nucleoproteínas/fisiologia , Proteínas Virais/genética , Proteínas Virais/fisiologia
13.
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
14.
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
15.
Biochim Biophys Acta ; 1814(9): 1154-61, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21570497

RESUMO

Trans-sialidases are surface-located proteins in Trypanosoma cruzi that participate in key parasite-host interactions and parasite virulence. These proteins are encoded by a large multigenic family, with tandem-repeated and individual genes dispersed throughout the genome. While a large number of genes encode for catalytically active enzyme isoforms, many others display mutations that involve catalytic residues. The latter ultimately code for catalytically inactive proteins with very high similarity to their active paralogs. These inactive members have been shown to be lectins, able to bind sialic acid and galactose in vitro, although their cellular functions are yet to be fully established. We now report structural and biochemical evidence extending the current molecular understanding of these lectins. We have solved the crystal structure of one such catalytically inactive trans-sialidase-like protein, after soaking with a specific carbohydrate ligand, sialyl-α2,3-lactose. Instead of the expected trisaccharide, the binding pocket was observed occupied by α-lactose, strongly suggesting that the protein retains residual hydrolytic activity. This hypothesis was validated by enzyme kinetics assays, in comparison to fully active wild-type trans-sialidase. Surface plasmon resonance also confirmed that these trans-sialidase-like lectins are not only able to bind small oligosaccharides, but also sialylated glycoproteins, which is relevant in the physiologic scenario of parasite infection. Inactive trans-sialidase proteins appear thus to be ß-methyl-galactosyl-specific lectins, evolved within an exo-sialidase scaffold, thus explaining why their lectin activity is triggered by the presence of terminal sialic acid.


Assuntos
Carboidratos/química , Glicoproteínas/química , Lectinas/química , Neuraminidase/química , Trypanosoma cruzi/enzimologia , Cristalografia por Raios X , Glicoproteínas/fisiologia , Hidrólise , Lactose/química , Modelos Moleculares , Neuraminidase/fisiologia , Estrutura Terciária de Proteína , alfa-Fetoproteínas/química
16.
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
17.
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
18.
Neurochem Res ; 37(6): 1170-84, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22407244

RESUMO

Twenty-five years ago the author proposed new ideas of glycoprotein (GPs) and glycosphingolipid (GSLs) functions at the cell membrane. The GPs, apart from their glycan carrying capacity, were assumed to have specific, protein associated, functions. In contrast, GSLs such as those of globo and neolacto/lacto series, were considered to be energetically cheap membrane packing substances, filling in membrane spaces not covered with functional GPs. The terminal carbohydrate structures of the neolacto/lacto GSLs, i.e., sialic acid residues and ABH glycotopes, were postulated to have either regulatory or protective functions, respectively. A special active role was ascribed to terminal ß-galactosyl residues of GSLs and GPs. Gangliosides were considered to be functional GSLs. In the present review the author discusses these old ideas in context of the contemporary knowledge and comes to the conclusion that they have not aged.


Assuntos
Sistema ABO de Grupos Sanguíneos/fisiologia , Membrana Celular/metabolismo , Glicoesfingolipídeos/fisiologia , Animais , Antígenos CD/fisiologia , Diferenciação Celular/fisiologia , Glicoproteínas/fisiologia , Humanos , Lactosilceramidas/fisiologia , Lectinas/fisiologia , Modelos Biológicos , Ácido N-Acetilneuramínico/fisiologia , Neuraminidase/fisiologia , Lectinas Semelhantes a Imunoglobulina de Ligação ao Ácido Siálico
19.
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
20.
J Virol ; 84(16): 8042-50, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20519385

RESUMO

The fitness of oseltamivir-resistant highly pathogenic H5N1 influenza viruses has important clinical implications. We generated recombinant human A/Vietnam/1203/04 (VN; clade 1) and A/Turkey/15/06 (TK; clade 2.2) influenza viruses containing the H274Y neuraminidase (NA) mutation, which confers resistance to NA inhibitors, and compared the fitness levels of the wild-type (WT) and resistant virus pairs in ferrets. The VN-H274Y and VN-WT viruses replicated to similar titers in the upper respiratory tract (URT) and caused comparable disease signs, and none of the animals survived. On days 1 to 3 postinoculation, disease signs caused by oseltamivir-resistant TK-H274Y virus were milder than those caused by TK-WT virus, and all animals survived. We then studied fitness by using a novel approach. We coinoculated ferrets with different ratios of oseltamivir-resistant and -sensitive H5N1 viruses and measured the proportion of clones in day-6 nasal washes that contained the H274Y NA mutation. Although the proportion of VN-H274Y clones increased consistently, that of TK-H274Y virus decreased. Mutations within NA catalytic (R292K) and framework (E119A/K, I222L, H274L, and N294S) sites or near the NA enzyme active site (V116I, I117T/V, Q136H, K150N, and A250T) emerged spontaneously (without drug pressure) in both pairs of viruses. The NA substitutions I254V and E276A could exert a compensatory effect on the fitness of VN-H274Y and TK-H274Y viruses. NA enzymatic function was reduced in both drug-resistant H5N1 viruses. These results show that the H274Y NA mutation affects the fitness of two H5N1 influenza viruses differently. Our novel method of assessing viral fitness accounts for both virus-host interactions and virus-virus interactions within the host.


Assuntos
Antivirais/farmacologia , Farmacorresistência Viral , Virus da Influenza A Subtipo H5N1/patogenicidade , Mutação de Sentido Incorreto , Neuraminidase/genética , Oseltamivir/farmacologia , Proteínas Virais/genética , Replicação Viral , Substituição de Aminoácidos/genética , Animais , Modelos Animais de Doenças , Furões , Virus da Influenza A Subtipo H5N1/efeitos dos fármacos , Virus da Influenza A Subtipo H5N1/genética , Masculino , Neuraminidase/fisiologia , Infecções por Orthomyxoviridae/patologia , Infecções por Orthomyxoviridae/virologia , Recombinação Genética , Sistema Respiratório/virologia , Análise de Sobrevida , Proteínas Virais/fisiologia , Virulência
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa