IMR Press / FBL / Volume 29 / Issue 4 / DOI: 10.31083/j.fbl2904164
Open Access Original Research
Infection of Human Macrophage-Like Cells by African Swine Fever Virus
Show Less
1 Laboratory of Cell Biology and Virology, Institute of Molecular Biology of NAS RA, 0014 Yerevan, Armenia
2 Laboratory of Molecular and Cellular Immunology, Institute of Molecular Biology NAS RA, 0014 Yerevan, Armenia
3 Experimental Laboratory, Yerevan State Medical University after M. Heratsi, 0025 Yerevan, Armenia
*Correspondence: zkaralyan@yahoo.com (Zaven A. Karalyan)
Front. Biosci. (Landmark Ed) 2024, 29(4), 164; https://doi.org/10.31083/j.fbl2904164
Submitted: 21 September 2023 | Revised: 15 January 2024 | Accepted: 18 February 2024 | Published: 23 April 2024
Copyright: © 2024 The Author(s). Published by IMR Press.
This is an open access article under the CC BY 4.0 license.
Abstract

Background: The African swine fever (ASF) virus (ASFV) and ASF-like viral sequences were identified in human samples and sewage as well as in different water environments. Pigs regularly experience infections by the ASFV. The considerable stability of the virus in the environment suggests that there is ongoing and long-term contact between humans and the ASFV. However, humans exhibit resistance to the ASFV, and the decisive factor in developing infection in the body is most likely the reaction of target macrophages to the virus. Therefore, this study aimed to characterize the responses of human macrophages to the virus and explore the distinct features of the viral replication cycle within human macrophages. Methods: The ASFV Armenia/07 strain was used in all experiments. In this study, quantitative real-time polymerase chain reaction (qRT-PCR) was used to determine the ASFV gene expression; flow cytometry analysis was performed to evaluate the effects of the inactive and active ASFV (inASFV and aASFV) treatments on the phenotype of THP-1-derived macrophages (Mφ0) and inflammatory markers. Moreover, other methods such as cell viability and apoptosis assays, staining techniques, phagocytosis assay, lysosome-associated membrane protein (LAMP-1) cytometry, and cytokine detection were used during experiments. Results: Our findings showed that the virus initiated replication by entering human macrophages. Subsequently, the virus shed its capsid and initiated the transcription of numerous viral genes, and at least some of these genes executed their functions. In THP-1-derived macrophages (Mφ0), the ASFV implemented several functions to suppress cell activity, although the timing of their implementation was slower compared with virus-sensitive porcine alveolar macrophages (PAMs). Additionally, the virus could not complete the entire replication cycle in human Mφ0, as indicated by the absence of viral factories and a decrease in infectious titers of the virus with each subsequent passage. Overall, the infection of Mφ0 with the ASFV caused significant alterations in their phenotype and functions, such as increased TLR2, TLR3, CD80, CD36, CD163, CXCR2, and surface LAMP-1 expression. Increased production of the tumor necrosis factor (TNF) and interleukin (IL)-10 and decreased production of interferon (IFN)-α were also observed. Taken together, the virus enters human THP-1-derived macrophages, starts transcription, and causes immunological responses by target cells but cannot complete the replicative cycle. Conclusion: These findings suggest that there may be molecular limitations within human macrophages that at least partially restrict the complete replication of the ASFV. Understanding the factors that hinder viral replication in Mφ0 can provide valuable insights into the host–virus interactions and the mechanisms underlying the resistance of human macrophages to the ASFV.

Keywords
African swine fever virus
THP-1-derived human macrophages
gene transcription
cytometry
TLRs
LAMP-1
Figures
Fig. 1.
Share
Back to top