Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
Gut Microbes ; 15(2): 2271620, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37953509

RESUMO

Microbiota are known to modulate the host response to influenza infection, but the mechanisms remain largely unknown. Gut metabolites are the key mediators through which gut microbes play anti-influenza effect. Transferring fecal metabolites from mice with high influenza resistance into antibiotic-treated recipient mice conferred resistance to influenza infections. By comparing the metabolites of different individuals with high or low influenza resistance, we identified and validated N-acetyl-D-glucosamine (GlcNAc) and adenosine showed strong positive correlations with influenza resistance and exerted anti-influenza effects in vivo or in vitro, respectively. Especially, GlcNAc mediated the anti-influenza effect by increasing the proportion and activity of NK cells. Several gut microbes, including Clostridium sp., Phocaeicola sartorii, and Akkermansia muciniphila, were positively correlated with influenza resistance, and can upregulate the level of GlcNAc in the mouse gut by exogenous supplementation. Subsequent studies confirmed that administering a combination of the three bacteria to mice via gavage resulted in similar modulation of NK cell responses as observed with GlcNAc. This study demonstrates that gut microbe-produced GlcNAc protects the host against influenza by regulating NK cells, facilitating the elucidation of the action mechanism of gut microbes mediating host influenza resistance.


Assuntos
Microbioma Gastrointestinal , Influenza Humana , Microbiota , Camundongos , Animais , Humanos , Células Matadoras Naturais , Fezes/microbiologia
2.
J Virol ; 97(10): e0092623, 2023 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-37754758

RESUMO

IMPORTANCE: Type I interferon (IFN-I), produced by the innate immune system, plays an essential role in host antiviral responses. Proper regulation of IFN-I production is required for the host to balance immune responses and prevent superfluous inflammation. IFN regulatory factor 3 (IRF3) and subsequent sensors are activated by RNA virus infection to induce IFN-I production. Therefore, proper regulation of IRF3 serves as an important way to control innate immunity and viral replication. Here, we first identified Prohibitin1 (PHB1) as a negative regulator of host IFN-I innate immune responses. Mechanistically, PHB1 inhibited the nucleus import of IRF3 by impairing its binding with importin subunit alpha-1 and importin subunit alpha-5. Our study demonstrates the mechanism by which PHB1 facilitates the replication of multiple RNA viruses and provides insights into the negative regulation of host immune responses.


Assuntos
Proteína DEAD-box 58 , Proibitinas , Vírus de RNA , Receptores Imunológicos , Transdução de Sinais , Replicação Viral , Proteína DEAD-box 58/antagonistas & inibidores , Proteína DEAD-box 58/metabolismo , Imunidade Inata , Fator Regulador 3 de Interferon/metabolismo , Carioferinas/metabolismo , Proibitinas/metabolismo , Receptores Imunológicos/antagonistas & inibidores , Receptores Imunológicos/metabolismo , Interferon Tipo I/biossíntese , Interferon Tipo I/imunologia , Vírus de RNA/crescimento & desenvolvimento , Vírus de RNA/imunologia , Vírus de RNA/metabolismo
3.
Mater Today Bio ; 16: 100444, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36204214

RESUMO

Accurate determination of the concentration and viability of the viral vaccine vectors is urgently needed for preventing the spread of the viral infections, but also supporting the development and assessment of recombinant virus-vectored vaccines. Herein, we describe a nanoplasmonic biosensor with nanoscale robot hand structure (Nano RHB) for the rapid, direct, and specific capture and quantification of adenovirus particles. The nanorobot allows simple operation in practical applications, such as real-time monitoring of vaccine quantity and quality, and evaluation of vaccine viability. Modification of the Nano RHB with branched gold nanostructures allow rapid and efficient assessment of human adenovirus viability, with ultrahigh detection sensitivity of only 100 copies/mL through one-step sandwich method. Nano RHB detection results were consistent with those from the gold standard median tissue culture infectious dose and real-time polymerase chain reaction assays. Additionally, the Nano RHB platform showed high detection specificity for different types of viral vectors and pseudoviruses. Altogether, these results demonstrate that the Nano RHB platform is a promising tool for efficient and ultrasensitive assessment of vaccines and gene delivery vectors.

4.
BMC Neurosci ; 23(1): 43, 2022 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-35794518

RESUMO

BACKGROUND: Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is highly contagious, and the neurological symptoms of SARS-CoV-2 infection have already been reported. However, the mechanisms underlying the effect of SARS-CoV-2 infection on patients with central nervous system injuries remain unclear. METHODS: The high-throughput RNA sequencing was applied to analyze the transcriptomic changes in SK-N-SH cells after SARS-CoV-2 infection. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analyses were performed to identify the functions of differentially expressed genes and related pathways. RESULTS: A total of 820 mRNAs were significantly altered, including 671 upregulated and 149 downregulated mRNAs (showing an increase of ≥ 2-fold or decrease to ≤ 0.5-fold, respectively; p ≤ 0.05). Moreover, we verified the significant induction of cytokines, chemokines, and their receptors, as well as the activation of NF-κB, p38, and Akt signaling pathways, in SK-N-SH by SARS-CoV-2. CONCLUSIONS: To our knowledge, this is the first time the transcriptional profiles of the host mRNAs involved in SARS-CoV-2 infection of SK-N-SH cells have been reported. These findings provide novel insight into the pathogenic mechanism of SARS-CoV-2 and might constitute a new approach for future prevention and treatment of SARS-CoV-2-induced central nervous system infection.


Assuntos
COVID-19 , Neuroblastoma , Citocinas , Humanos , NF-kappa B , RNA Mensageiro/metabolismo , SARS-CoV-2
5.
Microbiol Spectr ; 9(2): e0073421, 2021 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-34585989

RESUMO

The influenza A virus (IAV) infection is usually restricted to the respiratory tract and only rarely enters the central nervous system (CNS) and causes neurological symptoms. However, the roles of host factors involved in IAV infection in the CNS remain largely undetermined. Therefore, we aimed to characterize the host responses to IAV infection in the brain. We isolated a strain of IAV H5N6, which is neurotoxic and highly pathogenic to mice. High-throughput RNA sequencing (RNA-seq) revealed 240 differentially expressed genes in IAV-infected brains. Among the significantly downregulated genes, we focused on the gene encoding progesterone receptor membrane component-1 (PGRMC1) and observed that IAV H5N6 infection clearly inhibited PGRMC1 in both neuroblastoma and glioma cells. Furthermore, treatment with AG205, a PGRMC1-specific inhibitor, or PGRMC1 knockout promoted H5N6 multiplication in vitro, while overexpression of PGRMC1 resulted in opposite effects. Furthermore, AG205 treatment or PGRMC1 knockout significantly inhibited the retinoic acid-inducible gene I (RIG-I)-mediated interferon beta (IFN-ß) signaling pathway and reduced the levels of several antiviral proteins (Mx1 and ISG15). In addition, PGRMC1-mediated regulation of IFN signaling relied on inhibition of the expression and ubiquitination of RIG-I. The loss of PGRMC1 leads to an increased susceptibility of mice (brain and lung) to influenza A virus infection. Conclusively, our results show for the first time that IAV H5N6 downregulates PGRMC1 expression to contribute to virus proliferation by inhibiting RIG-I-mediated IFN-ß production in the brain. These findings may offer new insights regarding the interplay between IAV and host factors that may impact IAV pathogenicity in the brain. IMPORTANCE Central nervous system (CNS) disease is one of the most common extra-respiratory tract complications of influenza A virus (IAV) infections. However, there is still little knowledge about IAV regulating host responses in brain. In this study, we identified progesterone receptor membrane component-1 (PGRMC1) as a novel host factor involved in the replication and propagation of IAV H5N6 in the host brain. We also observed that PGRMC1 antagonism was required for viral evasion from the host immune response during IAV infection via inhibition of the retinoic acid-inducible gene I (RIG-I)-mediated interferon beta (IFN-ß) signaling pathway and downstream antiviral gene expression. This study revealed a newly identified regulatory mechanism used by IAV H5N6 to ensure its life cycle in the CNS.


Assuntos
Antivirais/farmacologia , Sistema Nervoso Central/metabolismo , Proteínas de Membrana/metabolismo , Receptores de Progesterona/metabolismo , Animais , Feminino , Interações Hospedeiro-Patógeno , Humanos , Vírus da Influenza A , Influenza Humana , Pulmão/virologia , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Infecções por Orthomyxoviridae , Receptores de Progesterona/genética , Transcriptoma , Replicação Viral
6.
EBioMedicine ; 70: 103505, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34332295

RESUMO

BACKGROUND: Staphylococcus aureus is a common human pathogen capable of causing diverse illnesses with possible recurrent infections. Although recent studies have highlighted the role of cellular immunity in recurrent infections, the mechanism by which S. aureus evades host responses remains largely unexplored. METHODS: This study utilizes in vitro and in vivo infection experiments to investigate difference of pro-inflammatory responses and subsequent adaptive immune responses between adsA mutant and WT S. aureus strain infection. FINDINGS: We demonstrated that adenosine synthase A (AdsA), a potent S. aureus virulence factor, can alter Th17 responses by interfering with NLRP3 inflammasome-mediated IL-1ß production. Specifically, S. aureus virulence factor AdsA dampens Th1/Th17 immunity by limiting the release of IL-1ß and other Th polarizing cytokines. In particular, AdsA obstructs the release of IL-1ß via the adenosine/A2aR/NLRP3 axis. Using a murine infection model, pharmacological inhibition of A2a receptor enhanced S. aureus-specific Th17 responses, whereas inhibition of NLRP3 and caspase-1 downregulated these responses. Our results showed that AdsA contributes to recurrent S. aureus infection by restraining protective Th1/Th17 responses. INTERPRETATION: Our study provides important mechanistic insights for therapeutic and vaccination strategies against S. aureus infections. FUNDING: This work was supported by grants from Shenzhen Peacock project (KQTD2015033-117210153), and Guangdong Science and Technology Department (2020B1212030004) to J.H. and China Postdoctoral Science Foundation (2019M663167) to BZZ. We also thank the L & T Charitable Foundation, the Guangdong Science and Technology Department (2020B1212030004), and the Program for Guangdong Introducing Innovative and Entrepreneurial Teams (2019BT02Y198) for their support.


Assuntos
Proteínas de Bactérias/imunologia , Infecções Estafilocócicas/imunologia , Staphylococcus aureus/enzimologia , Fatores de Virulência/imunologia , Adenosina/biossíntese , Animais , Células Cultivadas , Feminino , Humanos , Evasão da Resposta Imune , Interleucina-1beta/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Receptor A2A de Adenosina/metabolismo , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/imunologia , Staphylococcus aureus/patogenicidade , Células THP-1 , Células Th17/imunologia
7.
Front Vet Sci ; 8: 808234, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35127884

RESUMO

H5N6, the highly pathogenic avian influenza A virus (IAV) of clade 2.3.4.4, causes global outbreaks in poultry. H5N6 has become the dominant IAV subtype in waterfowls and causes human infections with high mortality rates. Here, we isolated two strains of H5N6, XGD and JX, from chickens and ducks, respectively. Growth kinetics were evaluated in duck embryo fibroblasts, chicken embryo fibroblasts, Madin-Darby canine kidney cells, and A549 lung carcinoma cells. Receptor binding specificity was analyzed via sialic acid-binding activity assay. The virulence of each strain was tested in BALB/c mice, and recombinant viruses were constructed via reverse genetics to further analyze the pathogenicity. The two strains showed no significant differences in growth kinetics in vitro; however, JX was more virulent in mice than XGD. We also identified 13 mutations in six viral proteins of the two strains through genetic analysis. Our study showed that the NS1 protein played a crucial role in enhancing the virulence of JX. Specifically, the amino acid 139D in NS1 contributed to the high pathogenicity. Therefore, 139D in NS1 might provide insight into the underlying mechanism of IAV adaptation in mammals.

8.
Genomics ; 113(1 Pt 2): 484-492, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32976974

RESUMO

N6-methyladenosine (m6A) mRNA methylation varies in response to stress. However, no map of m6A mRNA methylation has been obtained for sheep, nor is it known what effect this has on regulating heat stress in sheep. Here, we obtained m6A methylation maps of sheep liver tissues with and without heat stress by MeRIP-seq. In total, 8306 m6A peaks associated with 2697 genes were detected in the heat stress group, and 12,958 m6A peaks associated with 5494 genes were detected in the control group. Peaks were mainly enriched in coding regions and near stop codons with classical RRACH motifs. Methylation levels of heat stress and control sheep were higher near stop codons, although methylation was significantly lower in heat stress sheep. GO and KEGG revealed that differential m6A-containing genes were significantly enriched in the stress response and fat metabolism. Our results showed that m6A mRNA methylation modifications regulate heat stress in sheep.


Assuntos
Adenosina/análogos & derivados , Resposta ao Choque Térmico , Fígado/metabolismo , Processamento Pós-Transcricional do RNA , Adenosina/metabolismo , Animais , Metilação , RNA Mensageiro/metabolismo , Ovinos
9.
Front Immunol ; 11: 541267, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33123126

RESUMO

RIG-I and MDA5 are two key pattern recognition receptors that sense the invasion of RNA viruses and initiate type I interferon (IFN) response. Although these receptors are generally conserved in vertebrates, RIG-I is absent in chickens, whereas MDA5 is present. Chicken MDA5 (chMDA5) plays a pivotal role in sensing the invasion of RNA viruses into cells. However, unlike mammalian MDA5, where there are in-depth and extensive studies, regulation of the chMDA5-mediated signaling pathway remains unexplored. In this study, we performed a pulldown assay and mass spectrometry analysis to identify chicken proteins that could interact with the N terminal of chMDA5 (chMDA5-N) that contained two CARDs responsible for binding of the well-known downstream adaptor MAVS. We found that 337 host proteins could potentially interact with chMDA5-N, which were integrated to build a chMDA5-N-host association network and analyzed by KEGG pathway and Gene Ontology annotation. Results of our analysis revealed that diverse cellular processes, such as RNA binding and transport and protein translation, ribosome, chaperones, and proteasomes are critical cellular factors regulating the chMDA5-mediated signaling pathway. We cloned 64 chicken genes to investigate their effects on chMDA5-mediated chicken IFN-ß production and confirmed the association of chicken DDX5, HSPA8, HSP79, IFIT5, PRDX1, and hnRNPH2 with chMDA5-N. In particular, we found that chicken hnRNPH2 impairs the association between chMDA5-N and MAVS and thus acts as a check on the chMDA5-mediated signaling pathway. To our knowledge, this study is the first to analyze the chicken MDA5-host interactome, which provides fundamental but significant insights to further explore the mechanism of chicken MDA5 signaling regulation in detail.


Assuntos
Proteínas Aviárias/imunologia , Galinhas/imunologia , Ribonucleoproteínas Nucleares Heterogêneas Grupo F-H/imunologia , Interferon Tipo I/imunologia , Helicase IFIH1 Induzida por Interferon/imunologia , Animais , Proteínas Aviárias/genética , Linhagem Celular , Galinhas/genética , Ribonucleoproteínas Nucleares Heterogêneas Grupo F-H/genética , Interferon Tipo I/genética , Helicase IFIH1 Induzida por Interferon/genética
10.
J Virol ; 95(2)2020 12 22.
Artigo em Inglês | MEDLINE | ID: mdl-33087462

RESUMO

The viral ribonucleoprotein (vRNP) of the influenza A virus (IAV) is responsible for the viral RNA transcription and replication in the nucleus, and its functions rely on host factors. Previous studies have indicated that eukaryotic translation elongation factor 1 delta (eEF1D) may associate with RNP subunits, but its roles in IAV replication are unclear. Herein, we showed that eEF1D was an inhibitor of IAV replication because knockout of eEF1D resulted in a significant increase in virus yield. eEF1D interacted with RNP subunits polymerase acidic protein (PA), polymerase basic 1 (PB1), polymerase basic 2 (PB2), and also with nucleoprotein (NP) in an RNA-dependent manner. Further studies revealed that eEF1D impeded the nuclear import of NP and PA-PB1 heterodimer of IAV, thereby suppressing the vRNP assembly, viral polymerase activity, and viral RNA synthesis. Together, our studies demonstrate eEF1D negatively regulating the IAV replication by inhibition of the nuclear import of RNP subunits, which not only uncovers a novel role of eEF1D in IAV replication but also provides new insights into the mechanisms of nuclear import of vRNP proteins.IMPORTANCE Influenza A virus is the major cause of influenza, a respiratory disease in humans and animals. Different from most other RNA viruses, the transcription and replication of IAV occur in the cell nucleus. Therefore, the vRNPs must be imported into the nucleus for viral transcription and replication, which requires participation of host proteins. However, the mechanisms of the IAV-host interactions involved in nuclear import remain poorly understood. Here, we identified eEF1D as a novel inhibitor for the influenza virus life cycle. Importantly, eEF1D impaired the interaction between NP and importin α5 and the interaction between PB1 and RanBP5, which impeded the nuclear import of vRNP. Our studies not only reveal the molecular mechanisms of the nuclear import of IAV vRNP but also provide potential anti-influenza targets for antiviral development.


Assuntos
Núcleo Celular/metabolismo , Vírus da Influenza A/metabolismo , Proteínas do Nucleocapsídeo/metabolismo , Fator 1 de Elongação de Peptídeos/metabolismo , RNA Polimerase Dependente de RNA/metabolismo , Proteínas Virais/metabolismo , Células A549 , Transporte Ativo do Núcleo Celular , Células HEK293 , Humanos , Vírus da Influenza A/genética , Fator 1 de Elongação de Peptídeos/genética , Ligação Proteica , Multimerização Proteica , RNA Viral/biossíntese , RNA Polimerase Dependente de RNA/química , Transcrição Gênica , Proteínas do Core Viral/química , Proteínas do Core Viral/metabolismo , Proteínas Virais/química , Replicação Viral , alfa Carioferinas/metabolismo , beta Carioferinas/metabolismo
11.
Animals (Basel) ; 10(9)2020 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-32911823

RESUMO

Tibetan goat is an ancient breed, which inhabits the adverse conditions of the plateaus in China. To investigate the role of selection in shaping its genomes, we genotyped Tibetan goats (Nagqu Prefecture, above 4500 m) and three lowland populations (Xinjiang goats, Taihang goats and Huanghuai goats). The result of PCA, neighbor-joining (N-J) tree and model-based clustering showed that the genetic structure between the Tibetan goat and the three lowland populations has significant difference. As demonstrated by the di statistic, we found that some genes were related to the high-altitude adaptation of Tibetan goats. Functional analysis revealed that these genes were enriched in the VEGF (vascular endothelial growth factor) signaling pathway and melanoma, suggesting that nine genes (FGF2, EGFR, AKT1, PTEN, MITF, ENPEP, SIRT6, KDR, and CDC42) might have important roles in the high-altitude adaptation of Nagqu Tibetan goats. We also found that the LEPR gene was under the strongest selection (di value = 16.70), and it could induce upregulation of the hypoxic ventilatory response. In addition, five genes (LEPR, LDB1, EGFR, NOX4 and FGF2) with high di values were analyzed using q-PCR. Among them, we found that LEPR, LDB1 and FGF2 exhibited higher expression in the lungs of the Tibetan goats; LEPR, EGFR and LDB1 exhibited higher expression in the hearts of the Huanghuai goat. Our results suggest that LEPR, LDB1, EGFR and FGF2 genes may be related to the high-altitude adaptation of the goats. These findings improve our understanding of the selection of the high-altitude adaptability of the Nagqu Tibetan goats and provide new theoretical knowledge for the conservation and utilization of germplasm resources.

12.
Anal Chim Acta ; 1110: 19-25, 2020 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-32278394

RESUMO

Gold nanoparticles-based molecular beacon (Au NPs-MB), due to its extraordinary highly-quenching efficiency for fluorophores, has been extensively investigated and widely used for bioimaging and bioassay. However, apart from irreversible aggregation during the "aging" step, the preparation of Au NPs-MB often suffers from relatively poor salt stability, limiting its further in vivo application. Herein, Au NPs decorated magnetic microbeads was developed to construct a novel magnetic MB for DNA assay, which not only totally solved the aggregation problem of Au NPs, but also exhibited robust stability in buffer solution. Most importantly, the fluorescence signal of each microbeads could be collected individually, realizing single microbeads-based DNA imaging, and the detection limit for target DNA could reach 0.1 nM with the detection range of 0.2-20 nM. More importantly, because the magnetic microbeads with three sizes could be readily distinguished by flow cytometry, the employed three types of hairpin DNA probes can be labelled with the same dye FITC without fluorescence cross-interference. Therefore, multiplexing detection of tumor-suppressor genes (p16, p21 and p53) could be readily realized by using size-encoded magnetic microbeads pre-functionalized with corresponding probe DNA illustrating the potential of this method in multiplexing bioassay applications.


Assuntos
DNA/análise , Citometria de Fluxo , Ouro/química , Nanopartículas Metálicas/química , Humanos , Fenômenos Magnéticos , Tamanho da Partícula , Propriedades de Superfície
13.
Genes (Basel) ; 11(3)2020 03 13.
Artigo em Inglês | MEDLINE | ID: mdl-32183248

RESUMO

RIG-I and MDA5 are two key pattern recognition receptors that sense RNA virus invasion, but RIG-I is absent in chickens. Although chickens have intact MDA5, the genes downstream of chicken MDA5 (chMDA5) that may mediate antiviral response are not well studied. We compared the transcriptional profile of chicken embryonic fibroblasts (DF1) transfected with chMDA5, and poly(I:C), using RNA-seq. Transfected chMDA5 and poly(I:C) in DF1 cells were associated with the marked induction of many antiviral innate immune genes compared with control. Interestingly, nine interferon-stimulated genes (ISGs) were listed in the top 15 upregulated genes by chMDA5 and poly(I:C) transfection. We used real-time PCR to confirm the upregulation of the nine ISGs, namely, MX1, IFI6, IFIT5, RSAD2, OASL, CMPK2, HELZ2, EPSTI1, and OLFML1, by chMDA5 and poly(I:C) transfection in DF1 cells. However, avian influenza virus H5N6 infection only increased MX1, IFI6, IFIT5, RSAD2, and OASL expression levels. Further study showed that the overexpression of these five genes could significantly inhibit H5N6 virus replication. These results provide some insights into the gene expression pattern induced by chMDA5, which would be beneficial for understanding and identifying innate immune genes of chicken that may lead to new antiviral therapies.


Assuntos
Galinhas/genética , Helicase IFIH1 Induzida por Interferon/genética , Transcriptoma/genética , Animais , Embrião de Galinha , Galinhas/crescimento & desenvolvimento , Proteína DEAD-box 58/genética , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/genética , Imunidade Inata/genética , Interferons/genética , RNA-Seq , Receptores Imunológicos
14.
Front Vet Sci ; 7: 603584, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33409298

RESUMO

Avian influenza A (H7N9) viruses emerged in China in 2013 and caused a zoonotic disease associated with a high case-fatality ratio of more than 30%. Transcriptional profiles obtained using animal models reveal host responses to the disease, thereby providing insights into disease pathogenesis. Therefore, we aimed to characterize the host responses of the H7N9 virus infected-mouse lungs in this study. First, we isolated an avian-originated H7N9 strain, which was shown to be highly pathogenic to both chickens and mice. Genomic analysis results suggested that a 12-nucleotide-insertion was present at the hemagglutinin cleavage site, and both the hemagglutinin and neuraminidase genes belonged to the Yangtze River Delta lineage. RNA sequencing results revealed 566 differentially expressed genes in the H7N9-infected lungs. Moreover, transcriptome analysis revealed that over-activated antiviral signals and intense interferon-stimulated gene products possibly contributed to the high virulence of the virus in mice. Importantly, lung concentrations of inflammatory cytokines, including interleukin-1ß and interleukin-6, interferon-ß, and tumor necrosis factor-α, were upregulated in response to H7N9 virus infection. Overall, the present study provided a comprehensive understanding of H7N9 virus pathogenicity and correlated host immune responses.

15.
Autophagy ; 15(7): 1163-1181, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30741586

RESUMO

Influenza A virus can evade host innate immune response that is involved in several viral proteins with complicated mechanisms. To date, how influenza A M2 protein modulates the host innate immunity remains unclear. Herein, we showed that M2 protein colocalized and interacted with MAVS (mitochondrial antiviral signaling protein) on mitochondria, and positively regulated MAVS-mediated innate immunity. Further studies revealed that M2 induced reactive oxygen species (ROS) production that was required for activation of macroautophagy/autophagy and enhancement of MAVS signaling pathway. Importantly, the proton channel activity of M2 protein was demonstrated to be essential for ROS production and antagonizing the autophagy pathway to control MAVS aggregation, thereby enhancing MAVS signal activity. In conclusion, our studies provided novel insights into mechanisms of M2 protein in modulating host antiviral immunity and uncovered a new mechanism into biology and pathogenicity of influenza A virus. Abbreviations: AKT/PKB: AKT serine/threonine kinase; Apo: apocynin; ATG5: autophagy related 5; BAPTA-AM: 1,2-Bis(2-aminophenoxy) ethane-N,N,N',N'-tetraacetic acid tetrakis; BECN1: beclin 1; CARD: caspase recruitment domain; CCCP: carbonyl cyanide m-chlorophenylhydrazone; CQ: chloroquine; DCF: dichlorodihyd-rofluorescein; DPI: diphenyleneiodonium; DDX58: DExD/H-box helicase 58; eGFP: enhanced green fluorescent protein; EGTA: ethylene glycol-bis(2-aminoethylether)-N,N,N',N'-tetraacetic acid; ER: endoplasmic reticulum; hpi: hours post infection; IAV: influenza A virus; IFN: interferon; IP: immunoprecipitation; IRF3: interferon regulatory factor 3; ISRE: IFN-stimulated response elements; LIR: LC3-interacting region; MAP1LC3B/LC3B: microtubule associated protein 1 light chain 3 beta; MAVS: mitochondrial antiviral signaling protein; MMP: mitochondrial membrane potential; MOI, multiplicity of infection; mRFP: monomeric red fluorescent protein; MTOR: mechanistic target of rapamycin kinase; NC: negative control; NFKB/NF-κB: nuclear factor kappa B; PI3K: class I phosphoinositide 3-kinase; RLR: RIG-I-like-receptor; ROS: reactive oxygen species; SEV: sendai virus; TM: transmembrane; TMRM: tetramethylrhodamine methylester; VSV: vesicular stomatitis virus.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Imunidade Inata , Mitocôndrias/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Proteínas da Matriz Viral/metabolismo , Células A549 , Proteínas Adaptadoras de Transdução de Sinal/genética , Autofagossomos/imunologia , Autofagossomos/metabolismo , Autofagossomos/virologia , Autofagia/genética , Autofagia/imunologia , Proteína 5 Relacionada à Autofagia/genética , Proteína 5 Relacionada à Autofagia/metabolismo , Cálcio/imunologia , Cálcio/metabolismo , Células HEK293 , Células HeLa , Humanos , Imunidade Inata/genética , Vírus da Influenza A/imunologia , Vírus da Influenza A/metabolismo , Vírus da Influenza A/patogenicidade , Mitocôndrias/virologia , Dinâmica Mitocondrial/genética , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Ligação Proteica , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo , Proteínas da Matriz Viral/genética
16.
Cell Stress Chaperones ; 24(2): 333-342, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30701478

RESUMO

With the intensive development of the sheep industry and increasing global temperatures, heat stress in sheep has become an increasingly severe and important issue in recent years. The level of N6-methyladenosine (m6A) RNA methylation changes in response to stress plays important roles in stress responses. However, the role of m6A in the heat stress response of sheep remains unclear. To explore this issue, we measured heat stress protein (HSP) expression, liver function indexes, m6A on RNA, m6A-related enzyme expression, and tissue damage in sheep that had been subjected to heat stress. At the transcriptome level, our results showed significant increases in m6A on RNA and increased mRNA levels of HSPs (HSP70, HSP90, and HSP110) and m6A-related enzymes [METTL3 (methyltransferase-like 3), METTL14 (methyltransferase-like 14), WTAP (wilms tumor 1-associated protein), FTO (fat mass and obesity-associated protein), ALKBH5 (alkB homologue 5), YTHDF1-3 (YTH domain family proteins), and YTHDC1-2 (YTH domain-containing proteins)] following heat stress. At the protein level, the expression of METTL3, YTHDF1-2, and YTHDC2 showed no significant differences following heat stress. However, in contrast to its mRNA level after heat stress, the protein expression of YTHDF3 was reduced, while the expression of HSPs (HSP70, HSP90, and HSP110), METTL14, WTAP, FTO, ALKBH5, YTHDF3, and YTHDC1 increased in line with their measured mRNA levels. Histological experiments revealed that heat stress caused varying degrees of damage to sheep liver tissue. Moreover, immunohistochemical staining indicated that the m6A-related enzymes were expressed in sheep hepatocytes, and differences in expression patterns were observed between the control and heat stress groups. In summary, differences in the level of m6A and the expression of m6A-related enzymes in the liver of sheep were observed after heat stress. This indicates that m6A is involved in the regulation of heat stress in sheep. Our findings provide a new avenue for studying the responses to heat stress in sheep.


Assuntos
Proteínas de Choque Térmico/genética , Processamento Pós-Transcricional do RNA , RNA Mensageiro/metabolismo , Ovinos/genética , Ovinos/metabolismo , Adenosina/análogos & derivados , Homólogo AlkB 5 da RNA Desmetilase/genética , Animais , Resposta ao Choque Térmico , Metilação , Metiltransferases/genética , Proteínas Nucleares/genética , Proteínas de Ligação a RNA/genética
17.
Avian Pathol ; 48(3): 221-229, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30640510

RESUMO

Pasteurella multocida (P. multocida), a causative agent of fowl cholera, is an important pathogen in the poultry industry. In the present study, we found that the inactivated vaccine of P. multocida grown in an iron-restricted medium provided better protection than that grown in normal medium. Thus, we adopted a comparative proteomics approach, by using two-dimensional gel electrophoresis (2-DE), coupled with matrix-assisted laser desorption/ionization time of flight mass spectrometry (MALDI-TOF/TOF MS), to profile the supernatant proteins associated with P. multocida under both conditions. Eleven upregulated proteins were identified, including aspartate ammonia-lyase (AspA), diacylglycerol kinase (DgK), 30S ribosomal protein S6 (RpsF), and eight outer membrane proteins (OMPs). To further characterize the three novel supernatant proteins identified under iron-restricted conditions, the AspA, DgK and RpsF proteins were expressed and purified, and used as immunogens to vaccinate chickens. The results showed that AspA, DgK and RpsF proteins induced 80.0%, 66.7%, and 80.0% immunity, respectively. These data indicate that the three novel proteins identified in the supernatant of the culture media might play important roles in the survival of bacteria under iron-restricted conditions, and thus protect chickens against P. multocida. These findings also suggest that the proteins identified can be used as subunit vaccines.


Assuntos
Proteínas de Bactérias/imunologia , Vacinas Bacterianas/imunologia , Galinhas/imunologia , Cólera/prevenção & controle , Pasteurella multocida/metabolismo , Doenças das Aves Domésticas/prevenção & controle , Animais , Aspartato Amônia-Liase/imunologia , Cólera/imunologia , Diacilglicerol Quinase/imunologia , Ferro/metabolismo , Pasteurella multocida/genética , Pasteurella multocida/imunologia , Doenças das Aves Domésticas/imunologia , Proteômica , Proteínas Ribossômicas/imunologia , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz/veterinária , Vacinação/veterinária , Vacinas de Produtos Inativados/imunologia
18.
J Virol ; 93(4)2019 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-30541828

RESUMO

Influenza A virus (IAV) infection could induce autophagosome accumulation. However, the impact of the autophagy machinery on IAV infection remains controversial. Here, we showed that induction of cellular autophagy by starvation or rapamycin treatment increases progeny virus production, while disruption of autophagy using a small interfering RNA (siRNA) and pharmacological inhibitor reduces progeny virus production. Further studies revealed that alteration of autophagy significantly affects the early stages of the virus life cycle or viral RNA synthesis. Importantly, we demonstrated that overexpression of both the IAV M2 and NP proteins alone leads to the lipidation of LC3 to LC3-II and a redistribution of LC3 from the cytosol to punctate vesicles indicative of authentic autophagosomes. Intriguingly, both M2 and NP colocalize and interact with LC3 puncta during M2 or NP transfection alone and IAV infection, leading to an increase in viral ribonucleoprotein (vRNP) export and infectious viral particle formation, which indicates that the IAV-host autophagy interaction plays a critical role in regulating IAV replication. We showed that NP and M2 induce the AKT-mTOR-dependent autophagy pathway and an increase in HSP90AA1 expression. Finally, our studies provided evidence that IAV replication needs an autophagy pathway to enhance viral RNA synthesis via the interaction of PB2 and HSP90AA1 by modulating HSP90AA1 expression and the AKT-mTOR signaling pathway in host cells. Collectively, our studies uncover a new mechanism that NP- and M2-mediated autophagy functions in different stages of virus replication in the pathogenicity of influenza A virus.IMPORTANCE Autophagy impacts the replication cycle of many viruses. However, the role of the autophagy machinery in IAV replication remains unclear. Therefore, we explored the detailed mechanisms utilized by IAV to promote its replication. We demonstrated that IAV NP- and M2-mediated autophagy promotes IAV replication by regulating the AKT-mTOR signaling pathway and HSP90AA1 expression. The interaction of PB2 and HSP90AA1 results in the increase of viral RNA synthesis first; subsequently the binding of NP to LC3 favors vRNP export, and later the interaction of M2 and LC3 leads to an increase in the production of infectious viral particles, thus accelerating viral progeny production. These findings improve our understanding of IAV pathogenicity in host cells.


Assuntos
Autofagia/fisiologia , Vírus da Influenza A/metabolismo , Replicação Viral/fisiologia , Células A549 , Animais , Cães , Células HEK293 , Interações Hospedeiro-Patógeno , Humanos , Vírus da Influenza A/genética , Influenza Humana , Células Madin Darby de Rim Canino , Proteínas Associadas aos Microtúbulos/metabolismo , Ligação Proteica , RNA Interferente Pequeno/genética , RNA Viral/metabolismo , Ribonucleoproteínas/metabolismo , Transdução de Sinais , Sirolimo/farmacologia , Proteínas do Core Viral/metabolismo
19.
ACS Nano ; 13(1): 284-294, 2019 01 22.
Artigo em Inglês | MEDLINE | ID: mdl-30543399

RESUMO

Multifunctional nanoplatforms with special advantages in the diagnosis and treatment of cancer have been widely explored in nanomedicine. Herein, we synthesize two-dimensional core-shell nanocomposites (Ti3C2@Au) via a seed-growth method starting from the titanium carbide (Ti3C2) nanosheets, a classical type of MXene nanostructure. After growing gold on the surface of Ti3C2 nanosheets, the stability and biocompatibility of the nanocomposites are greatly improved by the thiol modification. Also importantly, the optical absorption in the near-infrared region is enhanced. Utilizing the ability of the high optical absorbance and strong X-ray attenuation, the synthesized Ti3C2@Au nanocomposites are used for photoacoustic and computed tomography dual-modal imaging. Importantly, the mild photothermal effect of the Ti3C2@Au nanocomposites could improve the tumor oxygenation, which significantly enhances the radiotherapy. No obvious long-term toxicity of the nanocomposites is found at the injected dose. This work highlights the promise of special properties of MXene-based multifunctional nanostructures for cancer theranostics.


Assuntos
Nanopartículas Metálicas/química , Imagem Multimodal/métodos , Nanocompostos/química , Neoplasias Experimentais/diagnóstico por imagem , Fototerapia/métodos , Radioterapia/métodos , Absorção de Radiação , Animais , Linhagem Celular Tumoral , Feminino , Ouro/química , Nanopartículas Metálicas/efeitos da radiação , Camundongos , Camundongos Endogâmicos BALB C , Nanocompostos/efeitos da radiação , Neoplasias Experimentais/terapia , Técnicas Fotoacústicas/métodos , Tomografia por Emissão de Pósitrons/métodos , Titânio/química
20.
Front Immunol ; 9: 409, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29599773

RESUMO

Tumor necrosis factor receptor-associated factor 3 (TRAF3), an intracellular signal transducer, is identified as an important component of Toll-like receptors and RIG-I-like receptors induced type I interferon (IFN) signaling pathways. Previous studies have clarified TRAF3 function in mammals, but little is known about the role of TRAF3 in ducks. Here, we cloned and characterized the full-length duck TRAF3 (duTRAF3) gene and an alternatively spliced isoform of duTRAF3 (duTRAF3-S) lacking the fragment encoding amino acids 217-319, from duck embryo fibroblasts (DEFs). We found that duTRAF3 and duTRAF3-S played different roles in regulating IFN-ß production in DEFs. duTRAF3 through its TRAF domain interacted with duMAVS or duTRIF, leading to the production of IFN-ß. However, duTRAF3-S, containing the TRAF domain, was unable to bind duMAVS or duTRIF due to the intramolecular binding between the N- and C-terminal of duTRAF3-S that blocked the function of its TRAF domain. Further analysis identified that duTRAF3-S competed with duTRAF3 itself for binding to duTRAF3, perturbing duTRAF3 self-association, which impaired the assembly of duTRAF3-duMAVS/duTRIF complex, ultimately resulted in a reduced production of IFN-ß. These findings suggest that duTRAF3 is an important regulator of duck innate immune signaling and reveal a novel mechanism for the negative regulation of IFN-ß production via changing the formation of the homo-oligomerization of wild molecules, implying a novel regulatory role of truncated proteins.


Assuntos
Proteínas Aviárias/metabolismo , Patos/imunologia , Fibroblastos/fisiologia , Interferon beta/metabolismo , Receptores do Fator de Necrose Tumoral/metabolismo , Fator 3 Associado a Receptor de TNF/metabolismo , Processamento Alternativo , Animais , Células Cultivadas , Clonagem Molecular , Imunidade Inata/genética , Multimerização Proteica/genética , Fator 3 Associado a Receptor de TNF/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA