• 제목/요약/키워드: Highly Pathogenic Avian Influenza Virus (HPAIV)

검색결과 10건 처리시간 0.035초

Identification of Differentially Expressed Genes in Ducks in Response to Avian Influenza A Virus Infections

  • Ndimukaga, Marc;Won, Kyunghye;Truong, Anh Duc;Song, Ki-Duk
    • 한국가금학회지
    • /
    • 제47권1호
    • /
    • pp.9-19
    • /
    • 2020
  • 본 연구는 고병원성 조류 인플루엔자 바이러스(high pathogenic avian influenza virus; HPAIV)와 저병원성 조류인플루엔자 바이러스(low pathogenic avian virus; LPAIV)가 감염된 오리의 폐세포에서 보고된 기존 전사체 데이터를 재분석하여 조류 인플루엔자 감염에 대응하는 숙주의 공통 전사체를 발굴하고, 생물정보 분석을 실시하여 바이오 마커로서 가능성을 제시하기 위하여 수행하였다. 이전 연구에서 생산된 microarray 데이터 세트를 재분석하여, HPAIV와 LPAIV가 각각 감염된 오리의 폐세포에서 각각 총 731 및 439개의 차등발현 유전자를 발굴하였다. 이들 차등발현 유전자 중에서, 227개의 유전자가 HPAIV와 LPAIV가 감염된 세포에서 공통적으로 조절되어, 193개의 유전자는 발현이 증가한 반면, 34개의 유전자는 발현이 감소하였다. 생물정보 분석을 통하여 차등발현 유전자들의 기능에 대한 주석달기를 실시하여, 리보솜과 단백질 대사 및 유전자 발현 관련 GO가 풍부해짐을 확인하였다. REACTOME 분석을 통하여 단백질 및 RNA 대사 경로 및 콜라겐 생합성과 변형을 포함한 조직 복구 경로가 조절됨을 확인하였다. 보다 구체적으로, 번역 및 RNA 품질 관리 경로에 관여하는 단백질을 코딩하는 유전자는 HPAIV 및 LPAIV 감염에 반응하여 발현의 증가 또는 감소하는 방향으로 조절되어 AIV가 숙주 번역 기계를 억제함으로써 숙주 방어 시스템을 회피할 수 있거나 번역을 위해 세포질로 내보내기 전에 AIV가 억제될 수 있음을 시사한다. AIV 감염은 바이러스 감염으로 인한 조직의 병변 형성을 조절하는 경로를 활성화시킬 수 있음을 시사한다.

Cytokine-cytokine receptor interactions in the highly pathogenic avian influenza H5N1 virus-infected lungs of genetically disparate Ri chicken lines

  • Vu, Thi Hao;Hong, Yeojin;Truong, Anh Duc;Lee, Jiae;Lee, Sooyeon;Song, Ki-Duk;Cha, Jihye;Dang, Hoang Vu;Tran, Ha Thi Thanh;Lillehoj, Hyun S.;Hong, Yeong Ho
    • Animal Bioscience
    • /
    • 제35권3호
    • /
    • pp.367-376
    • /
    • 2022
  • Objective: The highly pathogenic avian influenza virus (HPAIV) is a threat to the poultry industry as well as the economy and remains a potential source of pandemic infection in humans. Antiviral genes are considered a potential factor for HPAIV resistance. Therefore, in this study, we investigated gene expression related to cytokine-cytokine receptor interactions by comparing resistant and susceptible Ri chicken lines for avian influenza virus infection. Methods: Ri chickens of resistant (Mx/A; BF2/B21) and susceptible (Mx/G; BF2/B13) lines were selected by genotyping the Mx dynamin like GTPase (Mx) and major histocompatibility complex class I antigen BF2 genes. These chickens were then infected with influenza A virus subtype H5N1, and their lung tissues were collected for RNA sequencing. Results: In total, 972 differentially expressed genes (DEGs) were observed between resistant and susceptible Ri chickens, according to the gene ontology and Kyoto encyclopedia of genes and genomes pathways. In particular, DEGs associated with cytokine-cytokine receptor interactions were most abundant. The expression levels of cytokines (interleukin-1β [IL-1β], IL-6, IL-8, and IL-18), chemokines (C-C Motif chemokine ligand 4 [CCL4] and CCL17), interferons (IFN-γ), and IFN-stimulated genes (Mx1, CCL19, 2'-5'-oligoadenylate synthase-like, and protein kinase R) were higher in H5N1-resistant chickens than in H5N1-susceptible chickens. Conclusion: Resistant chickens show stronger immune responses and antiviral activity (cytokines, chemokines, and IFN-stimulated genes) than those of susceptible chickens against HPAIV infection.

Impact of inland waters on highly pathogenic avian influenza outbreaks in neighboring poultry farms in South Korea

  • Ahmad, Saleem;Koh, Kyeyoung;Yoo, Daesung;Suh, Gukhyun;Lee, Jaeil;Lee, Chang-Min
    • Journal of Veterinary Science
    • /
    • 제23권3호
    • /
    • pp.36.1-36.14
    • /
    • 2022
  • Background: Since 2003, the H5 highly pathogenic avian influenza (HPAI) subtype has caused massive economic losses in the poultry industry in South Korea. The role of inland water bodies in avian influenza (AI) outbreaks has not been investigated. Identifying water bodies that facilitate risk pathways leading to the incursion of the HPAI virus (HPAIV) into poultry farms is essential for implementing specific precautionary measures to prevent viral transmission. Objectives: This matched case-control study (1:4) examined whether inland waters were associated with a higher risk of AI outbreaks in the neighboring poultry farms. Methods: Rivers, irrigation canals, lakes, and ponds were considered inland water bodies. The cases and controls were chosen based on the matching criteria. The nearest possible farms located within a radius of 3 km of the case farms were chosen as the control farms. The poultry farms were selected randomly, and two HPAI epidemics (H5N8 [2014-2016] and H5N6 [2016-2017]) were studied. Conditional logistic regression analysis was applied. Results: Statistical analysis revealed that inland waters near poultry farms were significant risk factors for AI outbreaks. The study speculated that freely wandering wild waterfowl and small animals contaminate areas surrounding poultry farms. Conclusions: Pet birds and animals raised alongside poultry birds on farm premises may wander easily to nearby waters, potentially increasing the risk of AI infection in poultry farms. Mechanical transmission of the AI virus occurs when poultry farm workers or visitors come into contact with infected water bodies or their surroundings. To prevent AI outbreaks in the future, poultry farms should adopt strict precautions to avoid contact with nearby water bodies and their surroundings.

Analysis of miRNA expression in the trachea of Ri chicken infected with the highly pathogenic avian influenza H5N1 virus

  • Suyeon Kang;Thi Hao Vu;Jubi Heo;Chaeeun Kim;Hyun S. Lillehoj;Yeong Ho Hong
    • Journal of Veterinary Science
    • /
    • 제24권5호
    • /
    • pp.73.1-73.16
    • /
    • 2023
  • Background: Highly pathogenic avian influenza virus (HPAIV) is considered a global threat to both human health and the poultry industry. MicroRNAs (miRNA) can modulate the immune system by affecting gene expression patterns in HPAIV-infected chickens. Objectives: To gain further insights into the role of miRNAs in immune responses against H5N1 infection, as well as the development of strategies for breeding disease-resistant chickens, we characterized miRNA expression patterns in tracheal tissues from H5N1-infected Ri chickens. Methods: miRNAs expression was analyzed from two H5N1-infected Ri chicken lines using small RNA sequencing. The target genes of differentially expressed (DE) miRNAs were predicted using miRDB. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analysis were then conducted. Furthermore, using quantitative real-time polymerase chain reaction, we validated the expression levels of DE miRNAs (miR-22-3p, miR-146b-3p, miR27b-3p, miR-128-3p, miR-2188-5p, miR-451, miR-205a, miR-203a, miR-21-3p, and miR-200a3p) from all comparisons and their immune-related target genes. Results: A total of 53 miRNAs were significantly expressed in the infection samples of the resistant compared to the susceptible line. Network analyses between the DE miRNAs and target genes revealed that DE miRNAs may regulate the expression of target genes involved in the transforming growth factor-beta, mitogen-activated protein kinase, and Toll-like receptor signaling pathways, all of which are related to influenza A virus progression. Conclusions: Collectively, our results provided novel insights into the miRNA expression patterns of tracheal tissues from H5N1-infected Ri chickens. More importantly, our findings offer insights into the relationship between miRNA and immune-related target genes and the role of miRNA in HPAIV infections in chickens.

Reverse transcription loop-mediated isothermal amplification assay for the rapid and simultaneous detection of H5 and other subtypes of avian influenza viruses

  • Park, Yu-Ri;Kim, Eun-Mi;Han, Do-Hyun;Kang, Dae-Young;Yeo, Sang-Geon;Park, Choi-Kyu
    • 한국동물위생학회지
    • /
    • 제40권1호
    • /
    • pp.15-20
    • /
    • 2017
  • A two-tube reverse transcription loop-mediated isothermal amplification (RT-LAMP) assay was designed for the rapid visual detection of the M gene of all subtypes of avian influenza virus (AIV) and the H5 gene of the H5 subtype of highly pathogenic AIV (HPAIV). The reaction carried out in two tubes in a single step at $58^{\circ}C$ for 40 min, and the assay results could be visually detected by using hydroxynaphthol blue dye. Using M or H5 gene-specific primers, the assay successfully detected all subtypes or H5 subtypes of AIVs, including the Korean representative H5N1 and H5N8 HPAIVs. The detection limit of the assay was approximately $10^{2.0}$ $EID_{50}/reaction$ for the M and H5 genes of H5N1 HPAIV, respectively, and was more sensitive than that of previously reported RT-LAMP and comparable to that of real-time RT-PCR. These results suggest that the present RT-LAMP assay, with its high specificity, sensitivity, and simplicity, will be a useful diagnostic tool for surveillance of currently circulating H5 HPAIVs and other subtypes of AIV in bird population, even in under-equipped laboratories.

Molecular analysis of chicken interferon-alpha inducible protein 6 gene and transcriptional regulation

  • Jeong-Woong Park;Marc Ndimukaga;Jaerung So;Sujung Kim;Anh Duc Truong;Ha Thi Thanh Tran;Hoang Vu Dang;Ki-Duk Song
    • Journal of Animal Science and Technology
    • /
    • 제65권1호
    • /
    • pp.183-196
    • /
    • 2023
  • Interferon-alpha inducible protein 6 (IFI6) is an interferon-stimulated gene (ISG), belonging to the FAM14 family of proteins and is localized in the mitochondrial membrane, where it plays a role in apoptosis. Transcriptional regulation of this gene is poorly understood in the context of inflammation by intracellular nucleic acid-sensing receptors and pathological conditions caused by viral infection. In this study, chicken IFI6 (chIFI6) was identified and studied for its molecular features and transcriptional regulation in chicken cells and tissues, i.e., lungs, spleens, and tracheas from highly pathogenic avian influenza virus (HPAIV)-infected chickens. The chIFI6-coding sequences contained 1638 nucleotides encoding 107 amino acids in three exons, whereas the duck IFI6-coding sequences contained 495 nucleotides encoding 107 amino acids. IFI6 proteins from chickens, ducks, and quail contain an IF6/IF27-like superfamily domain. Expression of chIFI6 was higher in HPAIV-infected White Leghorn chicken lungs, spleens, and tracheas than in mock-infected controls. TLR3 signals regulate the transcription of chIFI6 in chicken DF-1 cells via the NF-κB and JNK signaling pathways, indicating that multiple signaling pathways differentially contribute to the transcription of chIFI6. Further research is needed to unravel the molecular mechanisms underlying IFI6 transcription, as well as the involvement of chIFI6 in the pathogenesis of HPAIV in chickens.

HPAI-resistant Ri chickens exhibit elevated antiviral immune-related gene expression

  • Thi Hao Vu;Jubi Heo;Yeojin Hong;Suyeon Kang;Ha Thi Thanh Tran;Hoang Vu Dang;Anh Duc Truong;Yeong Ho Hong
    • Journal of Veterinary Science
    • /
    • 제24권1호
    • /
    • pp.13.1-13.11
    • /
    • 2023
  • Background: Highly pathogenic avian influenza viruses (HPAIVs) is an extremely contagious and high mortality rates in chickens resulting in substantial economic impact on the poultry sector. Therefore, it is necessary to elucidate the pathogenic mechanism of HPAIV for infection control. Objective: Gene set enrichment analysis (GSEA) can effectively avoid the limitations of subjective screening for differential gene expression. Therefore, we performed GSEA to compare HPAI-infected resistant and susceptible Ri chicken lines. Methods: The Ri chickens Mx(A)/BF2(B21) were chosen as resistant, and the chickens Mx(G)/BF2(B13) were selected as susceptible by genotyping the Mx and BF2 genes. The tracheal tissues of HPAIV H5N1 infected chickens were collected for RNA sequencing followed by GSEA analysis to define gene subsets to elucidate the sequencing results. Results: We identified four differentially expressed pathways, which were immune-related pathways with a total of 78 genes. The expression levels of cytokines (IL-1β, IL-6, IL-12), chemokines (CCL4 and CCL5), type interferons and their receptors (IFN-β, IFNAR1, IFNAR2, and IFNGR1), Jak-STAT signaling pathway genes (STAT1, STAT2, and JAK1), MHC class I and II and their co-stimulatory molecules (CD80, CD86, CD40, DMB2, BLB2, and B2M), and interferon stimulated genes (EIF2AK2 and EIF2AK1) in resistant chickens were higher than those in susceptible chickens. Conclusions: Resistant Ri chickens exhibit a stronger antiviral response to HPAIV H5N1 compared with susceptible chickens. Our findings provide insights into the immune responses of genetically disparate chickens against HPAIV.

Antiviral Efficacy of Citra-kill®, Disinfectant Solution Against Avian Influenza Virus

  • Cha, Chun-Nam;Lee, Yeo-Eun;Kang, In-Jin;Yoo, Chang-Yeul;Park, Eun-Kee;An, Sun-Jeong;Kim, Suk;Lee, Hu-Jang
    • 한국식품위생안전성학회지
    • /
    • 제27권1호
    • /
    • pp.18-23
    • /
    • 2012
  • 본 연구에서는 4급 암모늄에 속하는 quaternary ammonium chloride와 구연산의 합제인 Citra-$Kill^{(R)}$의 조류인플루엔자에 대한 살바이러스 효과를 확인하기 위해 국립수의과학검역원의 소독제 효력시험 중 바이러스 소독제 효력시험에 따라 수행하였다. 소독제와 조류인플루엔자바이러스를 증류수, 경수, 그리고 유기물 조건에서 반응시킨 후, 중화액을 이용하여 중화시킨 다음, 중화된 용액 0.2 ml를 10일령의 계태아 요막에 주입하여 5일 동안 배양시킨 다음, 요막액을 채취하여 바이러스의 생존여부를 혈구응집반응을 통해 확인하여 소독제의 효력배수를 결정하였다. 본 연구의 결과, Citra-$Kill^{(R)}$은 증류수, 경수, 그리고 유기물 조건에서 각각 2,000, 1,500, 그리고 500배에서 조류인플루엔자바이러스를 불활성화시켜, 조류인플루엔자에 대해 뛰어난 소독효과를 갖고 있는 것이 확인되었다. 따라서 향후, 야외적용시험을 통해 실제적용에 따른 효과를 확인할 필요가 있을 것으로 사료된다.

The highly pathogenic H5N1 avian influenza virus induces the mitogen-activated protein kinase signaling pathway in the trachea of two Ri chicken lines

  • Vu, Thi Hao;Hong, Yeojin;Truong, Anh Duc;Lee, Sooyeon;Heo, Jubi;Lillehoj, Hyun S.;Hong, Yeong Ho
    • Animal Bioscience
    • /
    • 제35권7호
    • /
    • pp.964-974
    • /
    • 2022
  • Objective: The highly pathogenic avian influenza virus (HPAIV) is a threat to the poultry industry and economy and remains a potential source of pandemic infection in humans. Antiviral genes are considered a potential factor for studies on HPAIV resistance. Therefore, in this study, we investigated gene expression related to the mitogen-activated protein kinase (MAPK) signaling pathway by comparing non-infected, HPAI-infected resistant, and susceptible Ri chicken lines. Methods: Resistant (Mx/A; BF2/B21) and susceptible Ri chickens (Mx/G; BF2/B13) were selected by genotyping the Mx and BF2 genes. Then, the tracheal tissues of non-infected and HPAIV H5N1 infected chickens were collected for RNA sequencing. Results: A gene set overlapping test between the analyzed differentially expressed genes (DEGs) and functionally categorized genes was performed, including biological processes of the gene ontology (GO) and Kyoto encyclopedia of genes and genomes (KEGG) pathways. A total of 1,794 DEGs were observed between control and H5N1-infected resistant Ri chickens, 432 DEGs between control and infected susceptible Ri chickens, and 1,202 DEGs between infected susceptible and infected resistant Ri chickens. The expression levels of MAPK signaling pathway-related genes (including MyD88, NF-κB, AP-1, c-fos, Jun, JunD, MAX, c-Myc), cytokines (IL-1β, IL-6, IL-8), type I interferons (IFN-α, IFN-β), and IFN-stimulated genes (Mx1, CCL19, OASL, and PRK) were higher in H5N1-infected than in non-infected resistant Ri chickens. MyD88, Jun, JunD, MAX, cytokines, chemokines, IFNs, and IFN-stimulated expressed genes were higher in resistant-infected than in susceptible-infected Ri chickens. Conclusion: Resistant Ri chickens showed higher antiviral activity compared to susceptible Ri chickens, and H5N1-infected resistant Ri chickens had immune responses and antiviral activity (cytokines, chemokines, interferons, and IFN-stimulated genes), which may have been induced through the MAPK signaling pathway in response to H5N1 infection.

Genome-wide identification, organization, and expression profiles of the chicken fibroblast growth factor genes in public databases and Vietnamese indigenous Ri chickens against highly pathogenic avian influenza H5N1 virus infection

  • Anh Duc Truong;Ha Thi Thanh Tran;Nhu Thi Chu;Huyen Thi Nguyen;Thi Hao Vu;Yeojin Hong;Ki-Duk Song;Hoang Vu Dang;Yeong Ho Hong
    • Animal Bioscience
    • /
    • 제36권4호
    • /
    • pp.570-583
    • /
    • 2023
  • Objective: Fibroblast growth factors (FGFs) play critical roles in embryo development, and immune responses to infectious diseases. In this study, to investigate the roles of FGFs, we performed genome-wide identification, expression, and functional analyses of FGF family members in chickens. Methods: Chicken FGFs genes were identified and analyzed by using bioinformatics approach. Expression profiles and Hierarchical cluster analysis of the FGFs genes in different chicken tissues were obtained from the genome-wide RNA-seq. Results: A total of 20 FGF genes were identified in the chicken genome, which were classified into seven distinct groups (A-F) in the phylogenetic tree. Gene structure analysis revealed that members of the same clade had the same or similar exon-intron structure. Chromosome mapping suggested that FGF genes were widely dispersed across the chicken genome and were located on chromosomes 1, 4-6, 9-10, 13, 15, 28, and Z. In addition, the interactions among FGF proteins and between FGFs and mitogen-activated protein kinase (MAPK) proteins are limited, indicating that the remaining functions of FGF proteins should be further investigated in chickens. Kyoto encyclopedia of genes and genomes pathway analysis showed that FGF gene interacts with MAPK genes and are involved in stimulating signaling pathway and regulating immune responses. Furthermore, this study identified 15 differentially expressed genes (DEG) in 21 different growth stages during early chicken embryo development. RNA-sequencing data identified the DEG of FGFs on 1- and 3-days post infection in two indigenous Ri chicken lines infected with the highly pathogenic avian influenza virus H5N1 (HPAIV). Finally, all the genes examined through quantitative real-time polymerase chain reaction and RNA-Seq analyses showed similar responses to HPAIV infection in indigenous Ri chicken lines (R2 = 0.92-0.95, p<0.01). Conclusion: This study provides significant insights into the potential functions of FGFs in chickens, including the regulation of MAPK signaling pathways and the immune response of chickens to HPAIV infections.