Your browser doesn't support javascript.
loading
Transcriptome profiling in swine macrophages infected with African swine fever virus at single-cell resolution.
Zheng, Yuxuan; Li, Su; Li, Shi-Hua; Yu, Shaoxiong; Wang, Qihui; Zhang, Kehui; Qu, Liang; Sun, Yuan; Bi, Yuhai; Tang, Fuchou; Qiu, Hua-Ji; Gao, George F.
Afiliación
  • Zheng Y; CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Center for Influenza Research and Early-Warning (CASCIRE), Chinese Academy of Sciences (CAS), Beijing 100101, China.
  • Li S; State Key Laboratory of Veterinary Biotechnology, National High-Containment Laboratory for Animal Diseases Control and Prevention, and National African Swine Fever Para-Reference Laboratory, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China.
  • Li SH; CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Center for Influenza Research and Early-Warning (CASCIRE), Chinese Academy of Sciences (CAS), Beijing 100101, China.
  • Yu S; State Key Laboratory of Veterinary Biotechnology, National High-Containment Laboratory for Animal Diseases Control and Prevention, and National African Swine Fever Para-Reference Laboratory, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China.
  • Wang Q; CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Center for Influenza Research and Early-Warning (CASCIRE), Chinese Academy of Sciences (CAS), Beijing 100101, China.
  • Zhang K; Savaid Medical School, University of the Chinese Academy of Sciences, Beijing 100049, China.
  • Qu L; State Key Laboratory of Veterinary Biotechnology, National High-Containment Laboratory for Animal Diseases Control and Prevention, and National African Swine Fever Para-Reference Laboratory, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China.
  • Sun Y; State Key Laboratory of Veterinary Biotechnology, National High-Containment Laboratory for Animal Diseases Control and Prevention, and National African Swine Fever Para-Reference Laboratory, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China.
  • Bi Y; State Key Laboratory of Veterinary Biotechnology, National High-Containment Laboratory for Animal Diseases Control and Prevention, and National African Swine Fever Para-Reference Laboratory, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China.
  • Tang F; CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Center for Influenza Research and Early-Warning (CASCIRE), Chinese Academy of Sciences (CAS), Beijing 100101, China.
  • Qiu HJ; Savaid Medical School, University of the Chinese Academy of Sciences, Beijing 100049, China.
  • Gao GF; Beijing Advanced Innovation Center for Genomics (ICG), Peking University, Beijing 100871, China.
Proc Natl Acad Sci U S A ; 119(19): e2201288119, 2022 05 10.
Article en En | MEDLINE | ID: mdl-35507870
ABSTRACT
African swine fever virus (ASFV) is the causative agent of African swine fever, a highly contagious and usually fatal disease in pigs. The pathogenesis of ASFV infection has not been clearly elucidated. Here, we used single-cell RNA-sequencing technology to survey the transcriptomic landscape of ASFV-infected primary porcine alveolar macrophages. The temporal dynamic analysis of viral genes revealed increased expression of viral transmembrane genes. Molecular characteristics in the ASFV-exposed cells exhibited the activation of antiviral signaling pathways with increased expression levels of interferon-stimulated genes and inflammatory- and cytokine-related genes. By comparing infected cells with unexposed cells, we showed that the unfolded protein response (UPR) pathway was activated in low viral load cells, while the expression level of UPR-related genes in high viral load cells was less than that in unexposed cells. Cells infected with various viral loads showed signature transcriptomic changes at the median progression of infection. Within the infected cells, differential expression analysis and coregulated virus­host analysis both demonstrated that ASFV promoted metabolic pathways but inhibited interferon and UPR signaling, implying the regulation pathway of viral replication in host cells. Furthermore, our results revealed that the cell apoptosis pathway was activated upon ASFV infection. Mechanistically, the production of tumor necrosis factor alpha (TNF-α) induced by ASFV infection is necessary for cell apoptosis, highlighting the importance of TNF-α in ASFV pathogenesis. Collectively, the data provide insights into the comprehensive host responses and complex virus­host interactions during ASFV infection, which may instruct future research on antiviral strategies.
Asunto(s)
Palabras clave

Texto completo: 1 Bases de datos: MEDLINE Asunto principal: Fiebre Porcina Africana / Virus de la Fiebre Porcina Africana Límite: Animals Idioma: En Revista: Proc Natl Acad Sci U S A Año: 2022 Tipo del documento: Article País de afiliación: China

Texto completo: 1 Bases de datos: MEDLINE Asunto principal: Fiebre Porcina Africana / Virus de la Fiebre Porcina Africana Límite: Animals Idioma: En Revista: Proc Natl Acad Sci U S A Año: 2022 Tipo del documento: Article País de afiliación: China