Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.624
Filtrar
1.
Crit Rev Eukaryot Gene Expr ; 34(4): 45-54, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38505872

RESUMO

HDAC1 functions as an oncogene in multi-type cancers. This study aimed to investigate the roles of histone deacetylase 1 (HDAC1) in cervical cancer (CC). mRNA expression was determined using reverse transcription quantitative polymerase chain reaction. The protein-protein complexes was analyzed using co-immunoprecipitation assay. The binding sites between NRF2 and NEU1 were confirmed by chromatin immunoprecipitation assay. Cell viability was detected by CCK-8. Cell proliferation was measured using CCK-8 and colony formation assays. Cell migrative and invasive ability were determined using transwell assay. We found that HDAC1 was upregulated in CC patients and cells. Trichostatin A (TSA) treatment decreased the number of colonies and migrated and invaded cells. Moreover, HDAC1 interacted with NRF2 to downregulate NEU1 expression. NEU1 knockdown attenuated the effects of TSA and enhanced the aggressiveness of CC cells. In conclusion, HDAC1 functions as an oncogene in CC. Targeting HDAC1 may be an alternative strategy for CC.


Assuntos
Neoplasias do Colo do Útero , Feminino , Humanos , Regulação para Baixo , Neoplasias do Colo do Útero/genética , Histona Desacetilase 1/genética , Histona Desacetilase 1/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Sincalida/genética , Sincalida/metabolismo , Neuraminidase/genética , Neuraminidase/metabolismo
2.
Viruses ; 16(3)2024 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-38543754

RESUMO

The H274Y substitution (N2 numbering) in neuraminidase (NA) N1 confers oseltamivir resistance to A(H1N1) influenza viruses. This resistance has been associated with reduced N1 expression using transfected cells, but the effect of this substitution on the enzymatic properties and on the expression of other group-1-NA subtypes is unknown. The aim of the present study was to evaluate the antiviral resistance, enzymatic properties, and expression of wild-type (WT) and H274Y-substituted NA for each group-1-NA. To this end, viruses with WT or H274Y-substituted NA (N1pdm09 or avian N4, N5 or N8) were generated by reverse genetics, and for each reverse-genetic virus, antiviral susceptibility, NA affinity (Km), and maximum velocity (Vm) were measured. The enzymatic properties were coupled with NA quantification on concentrated reverse genetic viruses using mass spectrometry. The H274Y-NA substitution resulted in highly reduced inhibition by oseltamivir and normal inhibition by zanamivir and laninamivir. This resistance was associated with a reduced affinity for MUNANA substrate and a conserved Vm in all viruses. NA quantification was not significantly different between viruses carrying WT or H274Y-N1, N4 or N8, but was lower for viruses carrying H274Y-N5 compared to those carrying a WT-N5. In conclusion, the H274Y-NA substitution of different group-1-NAs systematically reduced their affinity for MUNANA substrate without a significant impact on NA Vm. The impact of the H274Y-NA substitution on viral NA expression was different according to the studied NA.


Assuntos
Vírus da Influenza A Subtipo H1N1 , Vírus da Influenza A , Influenza Humana , Humanos , Oseltamivir/farmacologia , Antivirais/farmacologia , Vírus da Influenza A/genética , Neuraminidase/genética , Neuraminidase/metabolismo , Vírus da Influenza A Subtipo H1N1/genética , Genética Reversa , Farmacorresistência Viral/genética , Substituição de Aminoácidos , Inibidores Enzimáticos/farmacologia
3.
Antiviral Res ; 224: 105853, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38430970

RESUMO

While clinical trials have illuminated both the virological and clinical efficacy of baloxavir for influenza and post-treatment viral resistance, these aspects warrant further study in real-world settings. In response, we executed a prospective, observational study of the Japanese 2022-2023 influenza season. A cohort of 73 A(H3N2)-diagnosed outpatients-36 treated with baloxavir, 20 with oseltamivir, and 17 with other neuraminidase inhibitors (NAIs)-were analyzed. Viral samples were collected before and after administering an antiviral on days 1, 5, and 10, respectively. Cultured viruses were amplified using RT-PCR and sequenced to detect mutations. Fever and other symptoms were tracked via self-reporting diaries. In the baloxavir cohort, viral detection was 11.1% (4/36) and 0% (0/36) on day 5 and day 10, respectively. Two isolates from day 5 (5.6%, 2/36) manifested I38T/M-substitutions in the polymerase acidic protein (PA). For oseltamivir and other NAIs, viral detection rates were 60.0% (12/20) and 52.9% (9/17) on day 5, and 16.7% (3/18) and 6.3% (1/16) on day 10, respectively. No oseltamivir-resistant neuraminidase mutations were identified after treatment. Median fever durations for the baloxavir, oseltamivir, and other NAI cohorts were 27.0, 38.0, and 36.0 h, respectively, with no significant difference. Two patients harboring PA I38T/M-substitutions did not exhibit prolonged fever or other symptoms. These findings affirm baloxavir's virological and clinical effectiveness against A(H3N2) in the 2022-2023 season and suggest limited clinical influence of post-treatment resistance emergence.


Assuntos
Dibenzotiepinas , Influenza Humana , Morfolinas , Triazinas , Humanos , Influenza Humana/tratamento farmacológico , Oseltamivir/uso terapêutico , Oseltamivir/farmacologia , Neuraminidase/genética , Neuraminidase/uso terapêutico , Vírus da Influenza A Subtipo H3N2/genética , Pacientes Ambulatoriais , Estações do Ano , Estudos Prospectivos , Antivirais/uso terapêutico , Antivirais/farmacologia , Piridonas/uso terapêutico , Inibidores Enzimáticos/farmacologia , Guanidinas/farmacologia , Febre/tratamento farmacológico
4.
Microb Pathog ; 190: 106628, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38508422

RESUMO

Rotavirus infections in suckling and weaning piglets cause severe dehydration and death, resulting in significant economic losses in the pig breeding industry. With the continuous emergence of porcine rotavirus (PoRV) variants and poor vaccine cross-protection among various genotypes, there is an urgent need to develop alternative strategies such as seeking effective antiviral products from nature, microbial metabolites and virus-host protein interaction. Sialidases play a crucial role in various physiopathological processes and offer a promising target for developing antivirus drugs. However, the effect of bacterial-derived sialidases on the infection of PoRVs remains largely unknown. Herein, we investigated the impact of bacterial-derived sialidases (sialidase Cp and Vc) on PoRV strain OSU(Group A) infection, using differentiated epithelial monkey kidney cells (MA104) as a model. Our results indicated that the pretreatment of MA104 with exogenous sialidases effectively suppressed PoRV OSU in a concentration-dependent manner. Notably, even at a concentration of 0.01 µU/mL, sialidases significantly inhibited the virus (MOI = 0.01). Meanwhile, we found that sialidase Vc pretreatment sharply reduced the binding rate of PoRV OSU. Last, we demonstrated that PoRV OSU might recognize α-2,3-linked sialic acid as the primary attachment factor in MA104. Our findings provide new insights into the underlying mechanism of PoRV OSU infections, shedding lights on the development of alternative antivirus approaches based on bacteria-virus interaction.


Assuntos
Neuraminidase , Infecções por Rotavirus , Rotavirus , Replicação Viral , Animais , Neuraminidase/metabolismo , Neuraminidase/genética , Rotavirus/efeitos dos fármacos , Rotavirus/fisiologia , Suínos , Replicação Viral/efeitos dos fármacos , Linhagem Celular , Células Epiteliais/virologia , Células Epiteliais/microbiologia , Ligação Viral/efeitos dos fármacos , Ácido N-Acetilneuramínico/metabolismo , Ácido N-Acetilneuramínico/farmacologia , Antivirais/farmacologia , Haplorrinos , Doenças dos Suínos/virologia , Doenças dos Suínos/microbiologia
5.
Respir Res ; 25(1): 134, 2024 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-38500102

RESUMO

Neu1 is a sialidase enzyme that plays a crucial role in the regulation of glycosylation in a variety of cellular processes, including cellular signaling and inflammation. In recent years, numerous evidence has suggested that human NEU1 is also involved in the pathogenesis of various respiratory diseases, including lung infection, chronic obstructive pulmonary disease (COPD), asthma, and pulmonary fibrosis. This review paper aims to provide an overview of the current research on human NEU1 and respiratory diseases.


Assuntos
Asma , Transtornos Respiratórios , Humanos , Neuraminidase/genética , Inflamação
6.
Hum Vaccin Immunother ; 20(1): 2304393, 2024 Dec 31.
Artigo em Inglês | MEDLINE | ID: mdl-38497413

RESUMO

Current influenza vaccines could be augmented by including recombinant neuraminidase (rNA) protein antigen to broaden protective immunity and improve efficacy. Toward this goal, we investigated formulation conditions to optimize rNA physicochemical stability. When rNA in sodium phosphate saline buffer (NaPBS) was frozen and thawed (F/T), the tetrameric structure transitioned from a "closed" to an "open" conformation, negatively impacting functional activity. Hydrogen deuterium exchange experiments identified differences in anchorage binding sites at the base of the open tetramer, offering a structural mechanistic explanation for the change in conformation and decreased functional activity. Change to the open configuration was triggered by the combined stresses of acidic pH and F/T. The desired closed conformation was preserved in a potassium phosphate buffer (KP), minimizing pH drop upon freezing and including 10% sucrose to control F/T stress. Stability was further evaluated in thermal stress studies where changes in conformation were readily detected by ELISA and size exclusion chromatography (SEC). Both tests were suitable indicators of stability and antigenicity and considered potential critical quality attributes (pCQAs). To understand longer-term stability, the pCQA profiles from thermally stressed rNA at 6 months were modeled to predict stability of at least 24-months at 5°C storage. In summary, a desired rNA closed tetramer was maintained by formulation selection and monitoring of pCQAs to produce a stable rNA vaccine candidate. The study highlights the importance of understanding and controlling vaccine protein structural and functional integrity.


Assuntos
Vacinas contra Influenza , Influenza Humana , Humanos , Influenza Humana/prevenção & controle , Neuraminidase/genética , Vacinas Sintéticas/genética , RNA
7.
Cell Rep Med ; 5(3): 101433, 2024 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-38401547

RESUMO

Inclusion of defined quantities of the two major surface proteins of influenza virus, hemagglutinin (HA) and neuraminidase (NA), could benefit seasonal influenza vaccines. Recombinant HA and NA multimeric proteins derived from three influenza serotypes, H1N1, H3N2, and type B, are surface displayed on nanoliposomes co-loaded with immunostimulatory adjuvants, generating "hexaplex" particles that are used to immunize mice. Protective immune responses to hexaplex liposomes involve functional antibody elicitation against each included antigen, comparable to vaccination with monovalent antigen particles. When compared to contemporary recombinant or adjuvanted influenza virus vaccines, hexaplex liposomes perform favorably in many areas, including antibody production, T cell activation, protection from lethal virus challenge, and protection following passive sera transfer. Based on these results, hexaplex liposomes warrant further investigation as an adjuvanted recombinant influenza vaccine formulation.


Assuntos
Vírus da Influenza A Subtipo H1N1 , Vacinas contra Influenza , Influenza Humana , Infecções por Orthomyxoviridae , Camundongos , Animais , Humanos , Hemaglutininas , Neuraminidase/genética , Vírus da Influenza A Subtipo H3N2 , Lipossomos , Adjuvantes Imunológicos , Vacinas Sintéticas
8.
Infect Immun ; 92(3): e0034423, 2024 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-38376159

RESUMO

As one of the keystone pathogens of periodontitis, the oral bacterium Porphyromonas gingivalis produces an array of virulence factors, including a recently identified sialidase (PG0352). Our previous report involving loss-of-function studies indicated that PG0352 plays an important role in the pathophysiology of P. gingivalis. However, this report had not been corroborated by gain-of-function studies or substantiated in different P. gingivalis strains. To fill these gaps, herein we first confirm the role of PG0352 in cell surface structures (e.g., capsule) and serum resistance using P. gingivalis W83 strain through genetic complementation and then recapitulate these studies using P. gingivalis ATCC33277 strain. We further investigate the role of PG0352 and its counterpart (PGN1608) in ATCC33277 in cell growth, biofilm formation, neutrophil killing, cell invasion, and P. gingivalis-induced inflammation. Our results indicate that PG0352 and PGN1608 are implicated in P. gingivalis cell surface structures, hydrophobicity, biofilm formation, resistance to complement and neutrophil killing, and host immune responses. Possible molecular mechanisms involved are also discussed. In summary, this report underscores the importance of sialidases in the pathophysiology of P. gingivalis and opens an avenue to elucidate their underlying molecular mechanisms.


Assuntos
Periodontite , Porphyromonas gingivalis , Humanos , Virulência , Neuraminidase/genética , Neuraminidase/metabolismo , Fatores de Virulência/genética , Fatores de Virulência/metabolismo , Periodontite/microbiologia
9.
mBio ; 15(2): e0220323, 2024 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-38206008

RESUMO

The ongoing transmission of influenza A viruses (IAV) for the past century continues to be a burden to humans. IAV binds terminal sialic acids (SA) of sugar molecules present within the upper respiratory tract (URT) in order to successfully infect hosts. The two most common SA structures that are important for IAV infection are those with α2,3- and α2,6-linkages. While mice were once considered to be an unsuitable system for studying IAV transmission due to their lack of α2,6-SA in the trachea, we have successfully demonstrated that IAV transmission in infant mice is remarkably efficient. This finding led us to re-evaluate the SA composition of the URT of mice using in situ immunofluorescence and examine its in vivo contribution to transmission for the first time. We demonstrate that mice express both α2,3- and α2,6-SA in the URT and that the difference in expression between infants and adults contributes to the variable transmission efficiencies observed. Furthermore, selectively blocking α2,3-SA or α2,6-SA within the URT of infant mice using lectins was necessary but insufficient at inhibiting transmission, and simultaneous blockade of both receptors was crucial in achieving the desired inhibitory effect. By employing a broadly acting neuraminidase to indiscriminately remove both SA moieties in vivo, we effectively suppressed viral shedding and halted the transmission of different strains of influenza viruses. These results emphasize the utility of the infant mouse model for studying IAV transmission and strongly indicate that broadly targeting host SA is an effective approach that inhibits IAV contagion.IMPORTANCEInfluenza virus transmission studies have historically focused on viral mutations that alter hemagglutinin binding to sialic acid (SA) receptors in vitro. However, SA binding preference does not fully account for the complexities of influenza A virus transmission in humans. Our previous findings reveal that viruses that are known to bind α2,6-SA in vitro have different transmission kinetics in vivo, suggesting that diverse SA interactions may occur during their life cycle. In this study, we examine the role of host SA on viral replication, shedding, and transmission in vivo. We highlight the critical role of SA presence during virus shedding, such that attachment to SA during virion egress is equally important as detachment from SA during virion release. These insights support the potential of broadly acting neuraminidases as therapeutic agents capable of restraining viral transmission in vivo. Our study unveils intricate virus-host interactions during shedding, highlighting the necessity to develop innovative strategies to effectively target transmission.


Assuntos
Vírus da Influenza A , Orthomyxoviridae , Humanos , Animais , Camundongos , Ácidos Siálicos/metabolismo , Traqueia , Neuraminidase/genética , Receptores Virais/metabolismo , Orthomyxoviridae/metabolismo
10.
Virol J ; 21(1): 7, 2024 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-38178138

RESUMO

BACKGROUND: Oncolytic viruses are being studied and developed as novel cancer treatments. Using directed evolution technology, structural modification of the viral surface protein increases the specificity of the oncolytic virus for a particular cancer cell. Newcastle disease virus (NDV) does not show specificity for certain types of cancer cells during infection; therefore, it has low cancer cell specificity. Hemagglutinin is an NDV receptor-binding protein on the cell surface that determines host cell tropism. NDV selectivity for specific cancer cells can be increased by artificial amino acid changes in hemagglutinin neuraminidase HN proteins via directed evolution, leading to improved therapeutic effects. METHODS: Sialic acid-binding sites (H domains) of the HN protein mutant library were generated using error-prone PCR. Variants of the H domain protein were screened by enzyme-linked immunosorbent assay using HCT 116 cancer cell surface molecules. The mutant S519G H domain protein showed the highest affinity for the surface protein of HCT 116 cells compared to that of different types of cancer cells. This showed that the S519G mutant H domain protein gene replaced the same part of the original HN protein gene, and S519G mutant recombinant NDV (rNDV) was constructed and recovered. S519G rNDV cancer cell killing effects were tested using the MTT assay with various cancer cell types, and the tumor suppression effect of the S519G mutant rNDV was tested in a xenograft mouse model implanted with cancer cells, including HCT 116 cells. RESULTS: S519G rNDV showed increased specificity and enhanced killing ability of HCT 116 cells among various cancer cells and a stronger suppressive effect on tumor growth than the original recombinant NDV. Directed evolution using an artificial amino acid change in the NDV HN (S519G mutant) protein increased its specificity and oncolytic effect in colorectal cancer without changing its virulence. CONCLUSION: These results provide a new methodology for the use of directed evolution technology for more effective oncolytic virus development.


Assuntos
Neoplasias Colorretais , Vírus Oncolíticos , Humanos , Animais , Camundongos , Vírus da Doença de Newcastle/genética , Vírus da Doença de Newcastle/metabolismo , Proteína HN/genética , Proteína HN/metabolismo , Neuraminidase/genética , Neuraminidase/metabolismo , Hemaglutininas , Ácido N-Acetilneuramínico/metabolismo , Células HCT116 , Vírus Oncolíticos/genética , Modelos Animais de Doenças , Proteínas de Membrana , Neoplasias Colorretais/terapia
11.
J Med Virol ; 96(2): e29427, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38288882

RESUMO

Influenza virus is known to cause mild to severe respiratory infections and is also prone to genetic mutations. Of all the mutations, neuraminidase (NA) gene mutations are a matter of concern, as most approved antivirals target this protein. During the 2020 influenza season, an emergence of mutation in the NA gene, affecting the binding of the World Health Organization (WHO)-recommended probes to the specific site of the NA gene, was reported by our group. As a result of this mutation, the WHO-recommended allelic discrimination real-time reverse transcriptase polymerase chain reaction (RT-PCR) assay was unable to detect wild-type (H275) or mutant oseltamivir-resistant (Y275) strains of influenza A(H1N1)pmd09 viruses. In the current study, the WHO-recommended probes were redesigned according to the mutation in the probe binding site. Fifty undetermined samples (2020-2021) from the previous study were retested with the newly designed probes and found to be positive for H275 and/or Y275. The results obtained were similar to the Sanger sequencing results from the previous study, suggesting that the redesigned probes were efficient in discriminating between wild-type and mutant-type viruses. Furthermore, 133 samples from 2022, making a total of 183 samples (2020-2022), were tested using improved allelic discrimination real-time RT-PCR, and the overall prevalence rate of oseltamivir resistance in 2020-2022 was found to be 0.54%.


Assuntos
Vírus da Influenza A Subtipo H1N1 , Influenza Humana , Humanos , Oseltamivir/farmacologia , Oseltamivir/uso terapêutico , Antivirais/farmacologia , Antivirais/uso terapêutico , Vírus da Influenza A Subtipo H1N1/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Mutação de Sentido Incorreto , Proteínas Virais/genética , Farmacorresistência Viral/genética , Mutação , Neuraminidase/genética
12.
Virology ; 590: 109954, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38086284

RESUMO

The possible emergence of drug-resistant avian flu raises concerns over the limited effectiveness of currently approved antivirals (neuraminidase inhibitors - NAIs) in the hypothetical event of a zoonotic spillover. Our study demonstrated that the recombinant avian A(H6N1) viruses showed reduced inhibition (RI) by multiple NAI drugs following the introduction of point mutations found predominantly in the neuraminidase gene (NA) of NAI-resistant human influenza strains (E119V, R292K and H274Y; N2 numbering). Moreover, A(H6N1)-H274Y showed increased replication efficiency in vitro, and a fitness advantage over wild-type (WT) when co-inoculated into embryonated hen's eggs. The results presented in our study together with the zoonotic potential of the A(H6N1) virus as evidenced by the human infection from 2013, highlight the need for enhanced monitoring of NAI resistance-associated signatures in circulating LPAI (low pathogenic avian influenza) globally.


Assuntos
Influenza Aviária , Influenza Humana , Animais , Feminino , Humanos , Oseltamivir/farmacologia , Galinhas , Neuraminidase/genética , Antivirais/farmacologia , Inibidores Enzimáticos/farmacologia , Mutação , Resistência a Medicamentos , Farmacorresistência Viral/genética
13.
Artigo em Inglês | MEDLINE | ID: mdl-38141291

RESUMO

Recombinant protein-based approaches are ideally suited for producing vaccine antigens that are not overly abundant in viruses, such as influenza neuraminidase (NA). However, obtaining sufficient quantities of recombinant viral surface antigens remains challenging, often resulting in the use of chimeric proteins with affinity tags that can invariably impact the antigen's properties. Here, we developed multistep chromatography approaches for purifying secreted recombinant NA (rNA) antigens that are derived from recent H1N1 and H3N2 viruses and produced using insect cells. Analytical analyses showed that these isolation procedures yielded homogenous tetrameric rNA preparations with consistent specific activities that were not possible from a common immobilized metal affinity chromatography purification procedure. The use of classical chromatography improved the rNA tetramer homogeneity by removing the requirement of the N-terminal poly-histidine affinity tag that was shown to promote higher order rNA oligomer formation. In addition, these procedures reduced the specific activity variation by eliminating the exposure to Ni2+ ions and imidazole, with the latter showing pH and NA subtype dependent effects. Together, these results demonstrate that purification by multistep chromatography improves the homogeneity of secreted rNAs and eliminates the need for affinity tag-based approaches that can potentially alter the properties of these recombinant antigens.


Assuntos
Vírus da Influenza A Subtipo H1N1 , Vacinas contra Influenza , Influenza Humana , Humanos , Neuraminidase/genética , Vírus da Influenza A Subtipo H1N1/genética , Vírus da Influenza A Subtipo H1N1/metabolismo , Vírus da Influenza A Subtipo H3N2/genética , Vírus da Influenza A Subtipo H3N2/metabolismo , Proteínas Recombinantes/genética , Cromatografia , RNA
14.
Zhonghua Yi Xue Za Zhi ; 103(41): 3287-3293, 2023 Nov 07.
Artigo em Chinês | MEDLINE | ID: mdl-37926573

RESUMO

Objective: To investigate the expression of neuraminidase-1 (NEU1) in Ewing sarcoma (ES) tissue and its effect on the proliferation and migration of ES cells. Methods: To obtain datasets of ES from the National Center for Biotechnology Information's High-Throughput Gene Expression Omnibus (GEO) for the analysis of NEU1 expression in ES; to acquire ES patient dataset from the International Cancer Genome Consortium (ICGC) database and apply Kaplan-Meier survival analysis to investigate the relationship between NEU1 and the prognosis of ES patients; adopting both univariate and multivariate Cox regression analysis to determine whether NEU1 is a prognostic factor for ES; adopting the Kyoto Encyclopedia of Genes and Genomes (KEGG) annotation to analyze the potential mechanism of NEU1 in regulating the malignant biological behavior of ES; adopting the real-time fluorescence quantitative polynucleotide chain reaction (RT-qPCR) to verify the expression of NEU1 in the human bone marrow mesenchymal stem cells (hBMSC) and the ES cell line RD-ES; adopting the transfection technology to knock down the expression of NEU1 in ES cell lines and divide them into two groups: shRNA-NEU1 and shRNA-NC to explore the effects of altered NEU1 expression on ES malignant behavior; adopting the cell counting kit (CCK-8) and cell clone formation experiment to detect the proliferation ability of two groups of cells; adopting the scratch healing experiment to test the cell migration ability of the two groups. Results: We retrieved and analyzed data from the GEO database, including GSE17674 (44 ES tissues and 18 normal tissues) and GSE17679 (87 ES tissues and 18 normal tissues), and found that NEU1 expression was significantly higher in ES tissues compared to normal control tissues (P<0.001). The complete gene expression and clinical information of 56 ES patients obtained from the ICGC database revealed that the ES patients with high NEU1 expression (n=28) had a significantly lower overall survival rates at different time points compared to those with low NEU1 expression (n=28) (HR=2.830, 95%CI:1.324-6.051, P=0.005). Univariate analysis indicated that NEU1 could impact ES patient prognosis (HR=1.049, 95%CI: 1.008-1.092, P=0.019), and multivariate analysis further suggested that NEU1 could serve as a risk factor for ES prognosis (HR=1.087, 95%CI: 1.028-1.148, P=0.003). KEGG results show that MAPK signaling pathway and cell adhesion molecule signaling pathway were potential mechanisms regulating the malignant process of ES. The RT-qPCR results showed that the expression level of NEU1 in the RD-ES cell line is significantly higher than that in the control cell hBMSC (2 184.23±527.32 vs 1.00±0.08, P<0.001). The CCK-8 experiment results show that the proliferation of RD-ES cells in the NEU1 knockdown group was lower than that in the control group at 24, 48, and 72 hours (0.494±0.126 vs 0.696±0.118, 0.657±0.096 vs 1.142±0.182, 1.053±0.064 vs 1.980±0.146, all P<0.001). The results of single cell clone formation experiment show that the number of colony formation in the low expression NEU1 group was significantly lower than that in the control group (184.2±123.9 vs 362.8±78.0, P=0.021). The cell scratch healing experiment finds that the average scratch distance of the NEU1 knockdown group was significantly lower than that of the control group (19.6%±5.7% vs 56.0%±7.6%, P<0.001). Conclusion: NEU1 may be a prognostic factor in ES, and its abnormal expression in ES can affect the proliferation and migration ability of the ES cells, leading to poor prognosis in ES patients.


Assuntos
Sarcoma de Ewing , Humanos , Proliferação de Células , Neuraminidase/genética , Neuraminidase/metabolismo , Prognóstico , RNA Interferente Pequeno , Sarcoma de Ewing/genética , Sarcoma de Ewing/patologia
15.
Sci Transl Med ; 15(724): eabp9599, 2023 11 29.
Artigo em Inglês | MEDLINE | ID: mdl-38019934

RESUMO

Epithelial cells are covered in carbohydrates (glycans). This glycan coat or "glycocalyx" interfaces directly with microbes, providing a protective barrier against potential pathogens. Bacterial vaginosis (BV) is a condition associated with adverse health outcomes in which bacteria reside in direct proximity to the vaginal epithelium. Some of these bacteria, including Gardnerella, produce glycosyl hydrolase enzymes. However, glycans of the human vaginal epithelial surface have not been studied in detail. Here, we elucidate key characteristics of the "normal" vaginal epithelial glycan landscape and analyze the impact of resident microbes on the surface glycocalyx. In human BV, glycocalyx staining was visibly diminished in electron micrographs compared to controls. Biochemical and mass spectrometric analysis showed that, compared to normal vaginal epithelial cells, BV cells were depleted of sialylated N- and O-glycans, with underlying galactose residues exposed on the surface. Treatment of primary epithelial cells from BV-negative women with recombinant Gardnerella sialidases generated BV-like glycan phenotypes. Exposure of cultured VK2 vaginal epithelial cells to recombinant Gardnerella sialidase led to desialylation of glycans and induction of pathways regulating cell death, differentiation, and inflammatory responses. These data provide evidence that vaginal epithelial cells exhibit an altered glycan landscape in BV and suggest that BV-associated glycosidic enzymes may lead to changes in epithelial gene transcription that promote cell turnover and regulate responses toward the resident microbiome.


Assuntos
Gardnerella vaginalis , Vaginose Bacteriana , Feminino , Humanos , Gardnerella vaginalis/genética , Gardnerella vaginalis/metabolismo , Vagina , Vaginose Bacteriana/genética , Vaginose Bacteriana/microbiologia , Bactérias/metabolismo , Polissacarídeos , Neuraminidase/genética , Neuraminidase/metabolismo
16.
J Lipid Res ; 64(12): 100463, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37871851

RESUMO

GM1 gangliosidosis is a neurodegenerative disorder caused by mutations in the GLB1 gene, which encodes lysosomal ß-galactosidase. The enzyme deficiency blocks GM1 ganglioside catabolism, leading to accumulation of GM1 ganglioside and asialo-GM1 ganglioside (GA1 glycolipid) in brain. This disease can present in varying degrees of severity, with the level of residual ß-galactosidase activity primarily determining the clinical course. Glb1 null mouse models, which completely lack ß-galactosidase expression, exhibit a less severe form of the disease than expected from the comparable deficiency in humans, suggesting a potential species difference in the GM1 ganglioside degradation pathway. We hypothesized this difference may involve the sialidase NEU3, which acts on GM1 ganglioside to produce GA1 glycolipid. To test this hypothesis, we generated Glb1/Neu3 double KO (DKO) mice. These mice had a significantly shorter lifespan, increased neurodegeneration, and more severe ataxia than Glb1 KO mice. Glb1/Neu3 DKO mouse brains exhibited an increased GM1 ganglioside to GA1 glycolipid ratio compared with Glb1 KO mice, indicating that NEU3 mediated GM1 ganglioside to GA1 glycolipid conversion in Glb1 KO mice. The expression of genes associated with neuroinflammation and glial responses were enhanced in Glb1/Neu3 DKO mice compared with Glb1 KO mice. Mouse NEU3 more efficiently converted GM1 ganglioside to GA1 glycolipid than human NEU3 did. Our findings highlight NEU3's role in ameliorating the consequences of Glb1 deletion in mice, provide insights into NEU3's differential effects between mice and humans in GM1 gangliosidosis, and offer a potential therapeutic approach for reducing toxic GM1 ganglioside accumulation in GM1 gangliosidosis patients.


Assuntos
Gangliosidose GM1 , Animais , Humanos , Camundongos , beta-Galactosidase/genética , beta-Galactosidase/metabolismo , beta-Galactosidase/uso terapêutico , Gangliosídeo G(M1)/metabolismo , Gangliosídeo G(M1)/uso terapêutico , Gangliosidose GM1/genética , Glicolipídeos , Neuraminidase/genética , Neuraminidase/uso terapêutico
17.
Vaccine ; 41(47): 6941-6951, 2023 11 13.
Artigo em Inglês | MEDLINE | ID: mdl-37884412

RESUMO

Influenza A virus in swine (IAV-S) continues to cause significant negative impact to both sows and growing pigs. The viral hemagglutinin (HA) and neuraminidase (NA) genes continue to evolve with HA diversifying at a faster rate than NA. Depending on country, whole inactivated virus (WIV) commercial and autogenous vaccines, as well as veterinary prescription vaccines targeting HA, are currently available. The use of these vaccines is focused on reducing virus and clinical signs in sows and to provide HA-specific maternally derived antibodies (MDA) to their suckling pigs. The deficiency in this strategy is that HA-MDA does not persist long enough to protect pigs through their growing phase from infection, and HA-MDA can interfere with effective pig immunization. This study evaluated the immunogenicity and efficacy of an adjuvanted, quadrivalent RNA Particle vaccine (Sequivity NA), currently licensed as Sequivity® IAV-S NA. This vaccine was formulated based on four NA antigens representing the major NA clades of IAV subtypes H1N1, H1N2 and H3N2 circulating in swine herds in the United States. In a series of trials, pigs were vaccinated twice, at three days and three weeks of age (WOA), followed by challenge with either homologous or heterologous IAV strains at 8 or 15 WOA. The Sequivity NA vaccine induced robust serum NA inhibition (NI) antibody and protected against IAV-S strains with homologous and heterologous NA to that of the vaccine. The magnitude and duration of nasal shedding was reduced in vaccinated-pigs challenged with either homologous or heterologous virus within the same NA clade. This NA-based RNA Particle vaccine avoids the known impact of HA-MDA on pig vaccination and provides a new tool to successfully reduce IAV-induced disease in the pig population.


Assuntos
Vírus da Influenza A Subtipo H1N1 , Vírus da Influenza A , Vacinas contra Influenza , Infecções por Orthomyxoviridae , Doenças dos Suínos , Suínos , Animais , Feminino , Infecções por Orthomyxoviridae/prevenção & controle , Infecções por Orthomyxoviridae/veterinária , Neuraminidase/genética , Vírus da Influenza A Subtipo H3N2 , Vacinas Combinadas , Vacinas contra Influenza/genética , Anticorpos , Anticorpos Antivirais
18.
Comp Immunol Microbiol Infect Dis ; 102: 102076, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37804607

RESUMO

We developed a method to determine the sequences of hemagglutinin (HA) and neuraminidase (NA) from RNA extracted directly from wild bird fecal samples, using Nanopore Flongle. We determined the nucleotide sequences and subtypes of HA and NA in 16 and 15 samples respectively, using Flongle. The results of HA and NA subtyping determined by the conventional method were consistent with their subtypes determined by our method, thereby the applicability of this method in the identification of HA and NA subtypes. In addition, the homology between the HA fragments in this and the Sanger methods ranged from 98.5 % to 100 %. Compared with conventional PCR with the Sanger method, this method can easily determine HA and NA subtypes and sequences directly from the fecal samples. It is easier to implement and has lower running costs (USD100$) than other NGS-based methods, making it a useful tool for avian influenza surveillance in wild birds.


Assuntos
Vírus da Influenza A , Influenza Aviária , Sequenciamento por Nanoporos , Animais , Hemaglutininas , Neuraminidase/genética , Sequenciamento por Nanoporos/veterinária , Animais Selvagens , Vírus da Influenza A/genética , Aves , Influenza Aviária/epidemiologia , Fezes , Filogenia
19.
Vet Microbiol ; 285: 109872, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37690146

RESUMO

Vaccines are widely used to prevent Newcastle disease virus (NDV). Under the pressure of immunization, NDVs with mutations among epitopes of F and HN protein were isolated, which indicates that the efficiency of vaccine may decrease in terms of preventing emerged NDV. However, the lack of evidences to support whether these mutations contribute to antigenic mutation and immune escape in NDV leading to the controversy that the matched vaccine is more effective than the mismatched vaccine. In this study, a genotype VII velogenic NDV strain (C22) was isolated from a vaccinated farm in Tibet, China. We found that this strain was close to NDV from east China, but it had a specific mutation (K138R) in one epitope (131DYIGGIGKE139) of HN protein. This mutation might change the interaction between amino acids in stalk-head link region of HN protein and then induce the specific antibody to worse recognize the C22 strain, but it did not alter viral virulence and growth ability. Then, the C22 strain was attenuated via modification of the F protein cleavage site to generate a matched vaccine. Comparing to a mismatched vaccine (LaSota), this matched vaccine showed advantages in inhibiting viral shedding and tissue damage. However, both vaccines induced chicken to generate similar level of neutralizing antibodies against C22, C22mut (R138K) and LaSota. These results suggest that the epitope mutation is insufficient to help NDV escaping neutralizing antibodies of vaccinated chicken, supporting that the merits of NDV matched vaccine are not totally related to humoral immunity.


Assuntos
Doença de Newcastle , Vacinas Virais , Animais , Vírus da Doença de Newcastle , Hemaglutininas/genética , Neuraminidase/genética , Tibet , Proteína HN/genética , Vacinas Virais/genética , Galinhas , Proteínas Virais/genética , Anticorpos Neutralizantes/genética , China , Variação Antigênica , Epitopos/genética , Anticorpos Antivirais , Genótipo
20.
Antiviral Res ; 218: 105719, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37717821

RESUMO

Influenza virus neuraminidase (NA) can act as a receptor-binding protein, a role commonly attributed to hemagglutinin (HA). In influenza A(H3N2) viruses, three NA amino acid residues have previously been associated with NA-mediated hemagglutination: T148, D151, and more recently, H150. These residues are part of the 150-loop of the NA monomer. Substitutions at 148 and 151 arise from virus propagation in laboratory cell cultures, whereas changes at 150 occurred during virus evolution in the human host. In this study, we examined the effect of natural amino acid polymorphism at position 150 on NA-mediated hemagglutination. Using the A/Puerto Rico/8/34 backbone, we generated a comprehensive panel of recombinant A(H3N2) viruses that have different NAs but shared an HA that displays poor binding to red blood cells (RBCs). None of the tested substitutions at 150 (C, H, L, R, and S) promoted NA-binding. However, we identified two new determinants of NA-binding, Q136K and T439R, that emerged during virus culturing. Similar to T148I, both Q136K and T439R reduced NA enzyme activity by 48-86% and inhibition (14- to 173-fold) by the NA inhibitor zanamivir. NA-binding was observed when a virus preparation contained approximately 10% of NA variants with either T148I or T439R, highlighting the benefit of using deep sequencing in virus characterization. Taken together, our findings provide new insights into the molecular mechanisms underlying the ability of NA to function as a binding protein. Information gained may aid in the design of new and improved NA-targeting antivirals.


Assuntos
Hemaglutinação , Vírus da Influenza A Subtipo H3N2 , Neuraminidase , Humanos , Aminoácidos/farmacologia , Antivirais/farmacologia , Neuraminidase/genética , Neuraminidase/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...