Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Publication year range
1.
Virol J ; 21(1): 4, 2024 01 04.
Article in English | MEDLINE | ID: mdl-38178220

ABSTRACT

BACKGROUND: Cross-species transmission of zoonotic IAVs to humans is potentially widespread and lethal, posing a great threat to human health, and their cross-species transmission mechanism has attracted much attention. miRNAs have been shown to be involved in the regulation of IAVs infection and immunity, however, few studies have focused on the molecular mechanisms underlying miRNAs and mRNAs expression after IAVs cross-species infection. METHODS: We used tree shrews, a close relative of primates, as a model and used RNA-Seq and bioinformatics tools to analyze the expression profiles of DEMs and DEGs in the nasal turbinate tissue at different time points after the newly emerged swine influenza A virus SW2783 cross-species infection with tree shrews, and miRNA-mRNA interaction maps were constructed and verified by RT-qPCR, miRNA transfection and luciferase reporter assay. RESULTS: 14 DEMs were screened based on functional analysis and interaction map, miR-760-3p, miR-449b-2, miR-30e-3p, and miR-429 were involved in the signal transduction process of replication and proliferation after infection, miR-324-3p, miR-1301-1, miR-103-1, miR-134-5p, miR-29a, miR-31, miR-16b, miR-34a, and miR-125b participate in negative feedback regulation of genes related to the immune function of the body to activate the antiviral immune response, and miR-106b-3p may be related to the cross-species infection potential of SW2783, and the expression level of these miRNAs varies in different days after infection. CONCLUSIONS: The miRNA regulatory networks were constructed and 14 DEMs were identified, some of them can affect the replication and proliferation of viruses by regulating signal transduction, while others can play an antiviral role by regulating the immune response. It indicates that abnormal expression of miRNAs plays a crucial role in the regulation of cross-species IAVs infection, which lays a solid foundation for further exploration of the molecular regulatory mechanism of miRNAs in IAVs cross-species infection and anti-influenza virus targets.


Subject(s)
MicroRNAs , Animals , Humans , Swine , MicroRNAs/genetics , MicroRNAs/metabolism , Influenza A Virus, H3N2 Subtype/genetics , Tupaia , Gene Expression Profiling , Tupaiidae/genetics , Shrews , RNA, Messenger
2.
Virus Res ; 324: 199027, 2023 01 15.
Article in English | MEDLINE | ID: mdl-36543317

ABSTRACT

Animal influenza viruses can spread across species and pose a fatal threat to human health due to the high pathogenicity and mortality. Animal models are crucial for studying cross-species infection and the pathogenesis of influenza viruses. Tupaia belangeri (tree shrew) has been emerging as an animal model for multiple human virus infections recently because of the close genetic relationship and phylogeny with humans. So far, tree shrew has been reported to be susceptible to human influenza virus subtype H1N1, avian influenza viruses subtype H9N2, subtype H5N1, and subtype H7N9. However, the pathogenicity, infection, and immunity of swine and land avian influenza viruses with low pathogenicity and the potential to jump to humans remain largely unexplored in the tree shrew model. Previously, our team has successfully isolated the newly emerging swine influenza virus subtype H3N2 (A/Swine/GX/NS2783/2010, SW2783) and avian influenza virus subtype H6N6 (A/CK/ZZ/346/2014, ZZ346). In this study, we observed the pathogenicity, immune characteristics, and cross-species infection potential ability of SW2783 and ZZ346 strains in tree shrew model with 50% tissue culture infective dose (TCID50), hematoxylin and eosin (HE) staining, immunohistochemistry (IHC), real-time quantitative PCR (qRT-PCR) and other experimental methods. Both animal-borne influenza viruses had a strong ability on tissue infection in the turbinate and the trachea of tree shrews in vitro, in which SW2783 showed stronger replication ability than in ZZ346. SW2783 and ZZ346 both showed pathogenic ability with infected tree shrews model in vivo without prior adaptive culture, which mainly happened in the upper respiratory tract. However, the infection ability was weak, the clinical symptoms were mild, and the histopathological changes in the respiratory tract were relatively light. Furthermore, innate immune responses and adaptive immunity were observed in the tree shrew model after the infection of SW2783 and ZZ346 strains. We observed that the unadapted SW2783 and ZZ346 virus could transmit among tree shrews by direct contact. We also observed that SW2783 virus could transmit from tree shrews to guinea pigs. These results indicated that both animal-borne influenza viruses could induce similar pathogenicity and immune response to those caused by human-common influenza viruses. Tree shrews may be an excellent animal model for studying the interaction between the influenza virus and the host and the cross-species infection mechanism of the animal influenza virus.


Subject(s)
Influenza A Virus, H1N1 Subtype , Influenza A Virus, H5N1 Subtype , Influenza A Virus, H7N9 Subtype , Influenza A Virus, H9N2 Subtype , Influenza, Human , Orthomyxoviridae Infections , Humans , Animals , Guinea Pigs , Tupaia , Tupaiidae , Influenza A Virus, H3N2 Subtype , Virulence , Shrews , Trachea/pathology , Virus Replication
3.
Virus Res ; 263: 34-46, 2019 04 02.
Article in English | MEDLINE | ID: mdl-30605755

ABSTRACT

Although several miRNAs have been demonstrated to be involved in the influenza virus replication cycle, the identification of miRNAs and mRNAs that are expressed in A549 cells infected with influenza A viruses (IAVs) from different host species has remained poorly studied. To investigate the molecular mechanisms associated with the differential expression of miRNAs during influenza A virus infection, we performed global miRNA and mRNA expression profiling in A549 cells infected with human-origin seasonal influenza A virus H3N2 (Human_Br07), swine-origin influenza A virus H1N1 (SW_3861) or avian-origin influenza A virus H3N2 (AVI_9990). The miRNA and mRNA expression profiles were obtained by microarray and high-throughput sequencing analyses, respectively. The integrated analysis of differentially expressed miRNAs (DEMs) and differentially expressed genes (DEGs) was performed using bioinformatics tools, and the expression of miRNAs and mRNAs was validated by real-time quantitative polymerase chain reaction (RT-qPCR). We identified 20 miRNAs (6 upregulated and 14 downregulated) and 1286 mRNAs (935 upregulated and 351 downregulated) exhibiting the same differential expression trends in three infected groups of cells compared with an uninfected control. An integrated analysis of these expression profiles identified 79 miRNA-mRNA pairs associated with the influenza A reference pathway, and 107 miRNA-mRNA interactions were correlated with the defense of the virus. Additionally, the obtained results were supported by an RT-qPCR analysis of 8 differentially expressed miRNAs (hsa-miR-210-3p, hsa-miR-296-5p, hsa-miR-371a-5p, hsa-miR-762, hsa-miR-937-5p, hsa-miR-1915-3p, hsa-miR-3665, and hsa-miR-1290) and 13 differentially expressed mRNAs (IFNL1, CXCL10, RSAD2, MX1, OAS2, IFIT2, IFI44 L, MX2, XAF1, NDRG1, FGA, EGLN3, and TFRC). Our findings indicate that dysregulated miRNA expression plays a crucial role in infection caused by IAVs originating from different species and provide a foundation for further investigations of the molecular regulatory mechanisms of miRNAs involved in influenza A virus infection.


Subject(s)
Epithelial Cells/pathology , Epithelial Cells/virology , Influenza A Virus, H1N1 Subtype/growth & development , Influenza A Virus, H3N2 Subtype/growth & development , MicroRNAs/analysis , RNA, Messenger/analysis , A549 Cells , Animals , Birds , Computational Biology , Gene Expression Profiling , High-Throughput Nucleotide Sequencing , Humans , Influenza A Virus, H1N1 Subtype/isolation & purification , Influenza A Virus, H3N2 Subtype/isolation & purification , Microarray Analysis , Real-Time Polymerase Chain Reaction , Swine
4.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 31(5): 599-604, 2015 May.
Article in Chinese | MEDLINE | ID: mdl-25940284

ABSTRACT

OBJECTIVE: To observe the effect of intraperitoneal injection of Newcastle disease virus (NDV) on tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) expression in mouse spleen NK cells and NK cells-mediated tumoricidal activity against mouse Novikoff hepatoma cell line, and explore the role of interferon (IFN)-γ in NDV-induced TRAIL expression and tumoricidal activity. METHODS: NDV was injected intraperitoneally to BALB/c mice and IFN-γ receptor-deficient (IFN-γR-/-) B6.129S7 mice. Twelve hours after injection, the concentration of IFN-γ in peripheral blood from BALB/c mice was determined by ELISA. Mouse spleen NK cells were separated. The mRNA and protein expression of TRAIL in NK cells were detected through reverse transcription PCR (RT-PCR) and Western blotting. Lactate dehydrogenase (LDH) release assay was used to determine the cytotoxic activity of NK cells against mouse hepatoma cells. RESULTS: NDV injection increased the IFN-γ concentration in peripheral blood of BALB/c mice, induced up-regulation of TRAIL at the mRNA and protein levels in mouse spleen NK cells, and enhanced the killing ability of mouse spleen NK cells towards Novikoff hepatoma cells. Blocking TRAIL by neutralizing antibody suppressed the cytotoxic activity of NK cells against Novikoff hepatoma cells. Furthermore, NDV injection in IFN-γR-/- B6.129S7 mice did not make significant difference from control group in TRAIL expression in spleen NK cells, and the tumoricidal activity of IFN-γR-/- B6.129S7 mouse spleen NK cells against Novikoff hepatoma cells was significantly lower than that of BALB/c mouse NK cells. CONCLUSION: Intraperitoneal injection with NDV could enhance tumoricidal activity of mouse spleen NK cells in vitro, and one of the mechanisms might be that NDV injection up-regulates TRAIL expression in NK cells through the IFN-γ receptor pathway.


Subject(s)
Carcinoma, Hepatocellular/pathology , Cytotoxicity, Immunologic , Killer Cells, Natural/immunology , Liver Neoplasms/pathology , Newcastle disease virus/physiology , TNF-Related Apoptosis-Inducing Ligand/genetics , Up-Regulation , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/virology , Interferon-gamma/blood , Liver Neoplasms/genetics , Liver Neoplasms/immunology , Liver Neoplasms/virology , Lymphocyte Activation , Mice , Rats , Spleen/immunology
5.
Anat Rec (Hoboken) ; 296(10): 1552-60, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23959779

ABSTRACT

Newcastle disease virus (NDV) is a potential antitumor agent, and its antitumor effect has been evaluated in preclinical tests. However, the mechanisms of NDV-based antitumor therapy are still not completely clear. In the present study we found that NDV-stimulation enhanced the killing ability of mouse spleen natural killer (NK) cells towards mouse hepatoma cell lines, and tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) plays an important role in this tumoricidal activity. NDV stimulation induced up-regulation of TRAIL both at the mRNA and protein levels in NK cells. Blocking TRAIL by antibody (Ab) almost completely eliminated the killing effect of NK cells on hepatoma cell lines. Furthermore, neutralizing interferon (IFN)-γ by Ab could inhibit TRAIL expression and tumoricidal activity of NDV-stimulated NK cells. These results indicated a substantial role of TRAIL as an effector molecule in NDV-induced NK cells mediated tumoricidal activity. The NDV stimulation triggered TRAIL expression in mouse spleen NK cells could be mediated by IFN-γ induction.


Subject(s)
Carcinoma, Hepatocellular/prevention & control , Cytotoxicity, Immunologic/immunology , Killer Cells, Natural/immunology , Liver Neoplasms/prevention & control , Newcastle disease virus/immunology , TNF-Related Apoptosis-Inducing Ligand/metabolism , Virus Replication/immunology , Animals , Antibodies, Monoclonal/pharmacology , Blotting, Western , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/virology , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Humans , In Vitro Techniques , Interferon-gamma/metabolism , Killer Cells, Natural/drug effects , Killer Cells, Natural/metabolism , Liver Neoplasms/immunology , Liver Neoplasms/virology , Mice , Mice, Inbred BALB C , Newcastle disease virus/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Spleen/drug effects , Spleen/immunology , Spleen/metabolism , TNF-Related Apoptosis-Inducing Ligand/antagonists & inhibitors , TNF-Related Apoptosis-Inducing Ligand/immunology , Tumor Cells, Cultured
7.
Zhonghua Zhong Liu Za Zhi ; 31(7): 490-4, 2009 Jul.
Article in Chinese | MEDLINE | ID: mdl-19950694

ABSTRACT

OBJECTIVE: To study the anti-tumor effects of Newcastle disease virus (NDV) strain D817 on human colon carcinoma model in nude mice. METHODS: The nude mouse model of human colon carcinoma was established by subcutaneous inoculation of human colon cancer LOVO cells. The tumor-bearing mice were given PBS, 5-Fu, high-dose NDV D817, moderate-dose NDV D817 or low-dose NDV D817 via caudal vein injection. The tumor size and weight of mice were measured. The liver damages were examined by histopathology. Apoptosis and necrosis of tumor cells were detected by flow cytometry. The endotumoral content of TNF-alpha was detected using a mouse TNF-alpha ELISA kit. The live virus was detected by hemagglutination (HA) test. RESULTS: The moderate-dose NDV D817 inhibited the tumor growth more apparently than 5-Fu. The tumor growth inhibition rate reached to 48.1%. The liver damage and the weight change caused by NDV were less severe. NDV D817 made an increased apoptosis index and induced production of TNF-alpha. Live virus was not detected in important organs except in the tumor of nude mice by HA test. CONCLUSION: In the anti-tumor process in nude mice bearing xenografts of human colon carcinoma, a suitable dose of NDV D817 is more safe and effective.


Subject(s)
Colonic Neoplasms/therapy , Newcastle disease virus/physiology , Animals , Apoptosis , Cell Line, Tumor , Colonic Neoplasms/pathology , Humans , Liver/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Random Allocation , Tumor Burden , Tumor Necrosis Factor-alpha/metabolism , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...