1
|
Durazo-Martinez K, Chaudhari J, Kumari S, Vu HLX. Porcine peritoneal macrophages are susceptible to porcine reproductive and respiratory syndrome virus infection. Front Microbiol 2024; 15:1505900. [PMID: 39723134 PMCID: PMC11668767 DOI: 10.3389/fmicb.2024.1505900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 11/18/2024] [Indexed: 12/28/2024] Open
Abstract
Previous studies have suggested that porcine peritoneal macrophages (PPMs) are resistant to PRRSV infection, whereas porcine alveolar macrophages (PAMs) are highly susceptible. This contrast is intriguing, as both cell types belong to the same monocyte/macrophage family. The current study aimed to investigate the host factors contributing to the differing susceptibility of PPMs and PAMs to PRRSV infection. We found that PPMs exhibit a higher frequency of CD14+ cells compared to PAMs, suggesting a more immature macrophage phenotype in PPMs. Importantly, PPMs expressed both CD163 and CD169, the key receptors for PRRSV entry, although the frequency and intensity of CD163 and CD169 expression were lower in PPMs than in PAMs. Despite these differences, PPMs were susceptible to both PRRSV-1 and PRRSV-2 isolates. Notably, PPMs susceptibility increased 10-fold when the cells were cultured for 1 day before infection. PRRSV infection in PPMs was dependent on CD163, as pretreatment with an anti-CD163 antibody significantly reduced infection. Overall, our results demonstrate that PPMs are susceptible to PRRSV infection, thereby expanding the understanding of PRRSV tropism.
Collapse
Affiliation(s)
- Kassandra Durazo-Martinez
- Department of Animal Science and Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE, United States
| | - Jayeshbhai Chaudhari
- Department of Animal Science and Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE, United States
| | - Sushmita Kumari
- School of Veterinary Science and Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE, United States
| | - Hiep L. X. Vu
- Department of Animal Science and Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE, United States
| |
Collapse
|
2
|
Zhu Z, Mao R, Liu B, Liu H, Shi Z, Zhang K, Liu H, Zhang D, Liu J, Zhao Z, Li K, Yang F, Cao W, Zhang X, Shen C, Sun D, Wang L, Tian H, Ru Y, Feng T, He J, Guo J, Zhang K, Tang Z, Zhang S, Ding C, Han J, Zheng H. Single-cell profiling of African swine fever virus disease in the pig spleen reveals viral and host dynamics. Proc Natl Acad Sci U S A 2024; 121:e2312150121. [PMID: 38412127 PMCID: PMC10927503 DOI: 10.1073/pnas.2312150121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Accepted: 01/08/2024] [Indexed: 02/29/2024] Open
Abstract
African swine fever, one of the major viral diseases of swine, poses an imminent threat to the global pig industry. The high-efficient replication of the causative agent African swine fever virus (ASFV) in various organs in pigs greatly contributes to the disease. However, how ASFV manipulates the cell population to drive high-efficient replication of the virus in vivo remains unclear. Here, we found that the spleen reveals the most severe pathological manifestation with the highest viral loads among various organs in pigs during ASFV infection. By using single-cell-RNA-sequencing technology and multiple methods, we determined that macrophages and monocytes are the major cell types infected by ASFV in the spleen, showing high viral-load heterogeneity. A rare subpopulation of immature monocytes represents the major population infected at late infection stage. ASFV causes massive death of macrophages, but shifts its infection into these monocytes which significantly arise after the infection. The apoptosis, interferon response, and antigen-presentation capacity are inhibited in these monocytes which benefits prolonged infection of ASFV in vivo. Until now, the role of immature monocytes as an important target by ASFV has been overlooked due to that they do not express classical monocyte marker CD14. The present study indicates that the shift of viral infection from macrophages to the immature monocytes is critical for maintaining prolonged ASFV infection in vivo. This study sheds light on ASFV tropism, replication, and infection dynamics, and elicited immune response, which may instruct future research on antiviral strategies.
Collapse
Affiliation(s)
- Zixiang Zhu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou730046, China
| | - Ruoqing Mao
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou730046, China
- African Swine Fever Regional Laboratory of China, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou730046, China
| | - Baohong Liu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou730046, China
| | - Huanan Liu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou730046, China
- African Swine Fever Regional Laboratory of China, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou730046, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou730046, China
| | - Zhengwang Shi
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou730046, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou730046, China
| | - Kunpeng Zhang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou730046, China
| | - Huisheng Liu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou730046, China
| | - Danyang Zhang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou730046, China
| | - Jia Liu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou730046, China
| | - Zhenxiang Zhao
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou730046, China
| | - Kangli Li
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou730046, China
| | - Fan Yang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou730046, China
| | - Weijun Cao
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou730046, China
| | - Xiangle Zhang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou730046, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou730046, China
| | - Chaochao Shen
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou730046, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou730046, China
| | - Dehui Sun
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou730046, China
| | - Liyuan Wang
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen518124, China
| | - Hong Tian
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou730046, China
| | - Yi Ru
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou730046, China
| | - Tao Feng
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou730046, China
| | - Jijun He
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou730046, China
- African Swine Fever Regional Laboratory of China, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou730046, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou730046, China
| | - Jianhong Guo
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou730046, China
- African Swine Fever Regional Laboratory of China, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou730046, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou730046, China
| | - Keshan Zhang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou730046, China
| | - Zhonglin Tang
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen518124, China
| | - Shilei Zhang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou730046, China
| | - Chan Ding
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai200241, China
| | - Jun Han
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing100193, China
| | - Haixue Zheng
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou730046, China
- African Swine Fever Regional Laboratory of China, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou730046, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou730046, China
| |
Collapse
|
3
|
Mu S, Chen L, Dong H, Li S, Zhang Y, Yin S, Tian Y, Ding Y, Sun S, Shang S, Guo H. Enhanced antigen-specific CD8 T cells contribute to early protection against FMDV through swine DC vaccination. J Virol 2024; 98:e0200223. [PMID: 38289108 PMCID: PMC10878267 DOI: 10.1128/jvi.02002-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 12/21/2023] [Indexed: 02/21/2024] Open
Abstract
Foot-and-mouth disease virus (FMDV) remains a challenge for cloven-hooved animals. The currently licensed FMDV vaccines induce neutralizing antibody (NAb)-mediated protection but show defects in the early protection. Dendritic cell (DC) vaccines have shown great potency in inducing rapid T-cell immunity in humans and mice. Whether DC vaccination could enhance early protection against FMDV has not been elaborately explored in domestic pigs. In this study, we employed DC vaccination as an experimental approach to study the roles of cellular immunity in the early protection against FMDV in pigs. Autologous DCs were differentiated from the periphery blood mononuclear cells of each pig, pulsed with inactivated FMDV (iFMDV-DC) and treated with LPS, and then injected into the original pigs. The cellular immune responses and protective efficacy elicited by the iFMDV-DC were examined by multicolor flow cytometry and tested by FMDV challenge. The results showed that autologous iFMDV-DC immunization induced predominantly FMDV-specific IFN-γ-producing CD4+ T cells and cytotoxic CD8+ T cells (CTLs), high NAb titers, compared to the inactivated FMDV vaccine, and accelerated the development of memory CD4 and CD8 T cells, which was concomitantly associated with early protection against FMDV virulent strain in pigs. Such early protection was associated with the rapid proliferation of secondary T-cell response after challenge and significantly contributed by secondary CD8 effector memory T cells. These results demonstrated that rapid induction of cellular immunity through DC immunization is important for improving early protection against FMDV. Enhancing cytotoxic CD8+ T cells may facilitate the development of more effective FMDV vaccines.IMPORTANCEAlthough the currently licensed FMDV vaccines provide NAb-mediated protection, they have defects in early immune protection, especially in pigs. In this study, we demonstrated that autologous swine DC immunization augmented the cellular immune response and induced an early protective response against FMDV in pigs. This approach induced predominantly FMDV-specific IFN-γ-producing CD4+ T cells and cytotoxic CD8+ T cells, high NAb titers, and rapid development of memory CD4 and CD8 T cells. Importantly, the early protection conferred by this DC immunization is more associated with secondary CD8+ T response rather than NAbs. Our findings highlighted the importance of enhancing cytotoxic CD8+ T cells in early protection to FMDV in addition to Th1 response and identifying a strategy or adjuvant comparable to the DC vaccine might be a future direction for improving the current FMDV vaccines.
Collapse
Affiliation(s)
- Suyu Mu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Lingbo Chen
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Hu Dong
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Shuai Li
- College of Veterinary Medicine, Institute of Comparative Medicine, Yangzhou University, Yangzhou, China
| | - Yun Zhang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Shuanghui Yin
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Yunfei Tian
- College of Veterinary Medicine, Institute of Comparative Medicine, Yangzhou University, Yangzhou, China
| | - Yaozhong Ding
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Shiqi Sun
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Shaobin Shang
- College of Veterinary Medicine, Institute of Comparative Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, China
| | - Huichen Guo
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| |
Collapse
|
4
|
Mehinagic K, Liniger M, Samoilenko M, Soltermann N, Gerber M, Ruggli N. A sensitive luciferase reporter assay for the detection of infectious African swine fever virus. J Virol Methods 2024; 323:114854. [PMID: 37989458 DOI: 10.1016/j.jviromet.2023.114854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 11/14/2023] [Accepted: 11/15/2023] [Indexed: 11/23/2023]
Abstract
African swine fever virus (ASFV) is a complex DNA virus causing severe hemorrhagic disease in domestic pigs and wild boar. The disease has spread worldwide, with important socio-economic consequences. Early virus detection and control measures are crucial as there are no effective vaccines nor antivirals on the market. While the diagnosis of ASFV is fast and based primarily on qPCR, the detection of infectious ASFV is a labor-intensive process requiring susceptible macrophages and subsequent antibody-based staining or hemadsorption. The latter cannot detect ASFV isolates devoid of functional CD2v (EP402R) expression. Here, we report the development of a plasmid-based reporter assay (RA) for the sensitive detection and titration of infectious ASFV. To this end, we constructed a plasmid for secreted NanoLuc luciferase (secNluc) expression driven by the ASFV DNA polymerase gene G1211R promoter. Infection of plasmid-transfected immortalized porcine kidney macrophages (IPKM) followed by measurement of secNluc from cell culture supernatants allowed reliable automated quantification of infectious ASFV. The RA-based titers matched the titers determined by conventional p72-staining or hemadsorption protocols. The novel assay is specific for ASFV as it does not detect classical swine fever virus nor porcine reproductive and respiratory syndrome virus. It is applicable to ASFV of different genotypes, virulence, and sources, including ASFV from sera and whole blood from infected pigs as well as non-hemadsorbing ASFV.
Collapse
Affiliation(s)
- Kemal Mehinagic
- Division of Virology, Institute of Virology and Immunology IVI, Mittelhäusern and Bern, Switzerland; Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Matthias Liniger
- Division of Virology, Institute of Virology and Immunology IVI, Mittelhäusern and Bern, Switzerland; Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Maksym Samoilenko
- Division of Virology, Institute of Virology and Immunology IVI, Mittelhäusern and Bern, Switzerland; Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Nick Soltermann
- Division of Virology, Institute of Virology and Immunology IVI, Mittelhäusern and Bern, Switzerland; Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Markus Gerber
- Division of Virology, Institute of Virology and Immunology IVI, Mittelhäusern and Bern, Switzerland; Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Nicolas Ruggli
- Division of Virology, Institute of Virology and Immunology IVI, Mittelhäusern and Bern, Switzerland; Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland.
| |
Collapse
|
5
|
Orosco FL. Host immune responses against African swine fever virus: Insights and challenges for vaccine development. Open Vet J 2023; 13:1517-1535. [PMID: 38292721 PMCID: PMC10824091 DOI: 10.5455/ovj.2023.v13.i12.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 11/22/2023] [Indexed: 02/01/2024] Open
Abstract
The African swine fever virus (ASFV) poses a serious threat to global swine populations, underscoring the urgent need for effective preventive strategies. This comprehensive review investigates the intricate interplay between innate, cellular, and humoral immunity against ASFV, with a focus on their relevance to vaccine development. By delving into immunopathogenesis and immunological challenges, this review article aims to provide a holistic perspective on the complexities of ASFV infections and immune evasion. Key findings underscore the critical role of innate immune recognition in shaping subsequent adaptive immune defenses, potential protective antigens, and the multifaceted nature of ASFV-specific antibodies and cytotoxic T-cell responses. Despite advancements, the unique attributes of ASFV present hurdles in the development of a successful vaccine. In conclusion, this review examines the current state of ASFV immune responses and offers insights into future research directions, fostering the development of effective interventions against this devastating pathogen.
Collapse
Affiliation(s)
- Fredmoore L. Orosco
- Virology and Vaccine Institute of the Philippines Program, Department of Science and Technology, Industrial Technology Development Institute, Taguig, Philippines
- S&T Fellows Program, Department of Science and Technology, Taguig, Philippines
- Department of Biology, College of Arts and Sciences, University of the Philippines Manila, Manila, Philippines
| |
Collapse
|
6
|
Reis AL, Rathakrishnan A, Goulding LV, Barber C, Goatley LC, Dixon LK. Deletion of the gene for the African swine fever virus BCL-2 family member A179L increases virus uptake and apoptosis but decreases virus spread in macrophages and reduces virulence in pigs. J Virol 2023; 97:e0110623. [PMID: 37796125 PMCID: PMC10617521 DOI: 10.1128/jvi.01106-23] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 08/23/2023] [Indexed: 10/06/2023] Open
Abstract
IMPORTANCE African swine fever virus (ASFV) causes a lethal disease of pigs with high economic impact in affected countries in Africa, Europe, and Asia. The virus encodes proteins that inhibit host antiviral defenses, including the type I interferon response. Host cells also activate cell death through a process called apoptosis to limit virus replication. We showed that the ASFV A179L protein, a BCL-2 family apoptosis inhibitor, is important in reducing apoptosis in infected cells since deletion of this gene increased cell death and reduced virus replication in cells infected with the A179L gene-deleted virus. Pigs immunized with the BeninΔA179L virus showed no clinical signs and a weak immune response but were not protected from infection with the deadly parental virus. The results show an important role for the A179L protein in virus replication in macrophages and virulence in pigs and suggest manipulation of apoptosis as a possible route to control infection.
Collapse
Affiliation(s)
| | | | | | - Claire Barber
- The Pirbright Institute, Woking, Surrey, United Kingdom
| | | | | |
Collapse
|
7
|
Takenouchi T, Masujin K, Suzuki S, Haraguchi S, Hiramatsu K, Kokuho T, Uenishi H. Establishment and characterization of the immortalized porcine lung-derived mononuclear phagocyte cell line. Front Vet Sci 2022; 9:1058124. [PMID: 36467652 PMCID: PMC9715978 DOI: 10.3389/fvets.2022.1058124] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 11/04/2022] [Indexed: 08/27/2023] Open
Abstract
Mononuclear phagocytes (MNP), including monocytes, dendritic cells (DC), and macrophages, play critical roles in innate immunity. MNP are abundant in the lungs and contribute to host defense against airborne agents and pulmonary immune homeostasis. In this study, we isolated porcine lung-derived MNP (PLuM) from primary cultures of parenchymal lung cells and then immortalized them by transferring the SV40 large T antigen gene and porcine telomerase reverse transcriptase gene using lentiviral vectors. The established cell line, immortalized PLuM (IPLuM), expressed DC/macrophage markers; i.e., CD163, CD172a, and major histocompatibility complex class II, whereas they did not express a porcine monocyte-specific marker, CD52. The expression patterns of these cell surface markers indicate that IPLuM originate from the DC/macrophage lineage rather than the monocyte lineage. The bacterial cell wall components muramyl dipeptide and lipopolysaccharide induced the production of the interleukin-1 family of pro-inflammatory cytokines in IPLuM. Phagocytotic activity was also detected by time-lapse fluorescence imaging of live cells when IPLuM were cultured in the presence of pHrodo dye-conjugated E. coli BioParticles. It is worth noting that IPLuM are susceptible to African swine fever virus infection and support the virus' efficient replication in vitro. Taken together, the IPLuM cell line may be a useful model for investigating host-agent interactions in the respiratory microenvironments of the porcine lung.
Collapse
Affiliation(s)
- Takato Takenouchi
- Institute of Agrobiological Sciences, National Agriculture and Food Research Organization, Tsukuba, Japan
| | - Kentaro Masujin
- Division of Transboundary Animal Disease Research, National Institute of Animal Health, National Agriculture and Food Research Organization, Tokyo, Japan
| | - Shunichi Suzuki
- Institute of Agrobiological Sciences, National Agriculture and Food Research Organization, Tsukuba, Japan
| | - Seiki Haraguchi
- Institute of Agrobiological Sciences, National Agriculture and Food Research Organization, Tsukuba, Japan
| | - Kanae Hiramatsu
- Institute of Agrobiological Sciences, National Agriculture and Food Research Organization, Tsukuba, Japan
- Division of Infectious Animal Disease Research, National Institute of Animal Health, National Agriculture and Food Research Organization, Tsukuba, Japan
| | - Takehiro Kokuho
- Division of Transboundary Animal Disease Research, National Institute of Animal Health, National Agriculture and Food Research Organization, Tokyo, Japan
| | - Hirohide Uenishi
- Institute of Agrobiological Sciences, National Agriculture and Food Research Organization, Tsukuba, Japan
| |
Collapse
|
8
|
Bosch-Camós L, Alonso U, Esteve-Codina A, Chang CY, Martín-Mur B, Accensi F, Muñoz M, Navas MJ, Dabad M, Vidal E, Pina-Pedrero S, Pleguezuelos P, Caratù G, Salas ML, Liu L, Bataklieva S, Gavrilov B, Rodríguez F, Argilaguet J. Cross-protection against African swine fever virus upon intranasal vaccination is associated with an adaptive-innate immune crosstalk. PLoS Pathog 2022; 18:e1010931. [PMID: 36350837 PMCID: PMC9645615 DOI: 10.1371/journal.ppat.1010931] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 10/17/2022] [Indexed: 11/11/2022] Open
Abstract
African swine fever virus (ASFV) is causing a worldwide pandemic affecting the porcine industry and leading to important global economic consequences. The virus causes a highly lethal hemorrhagic disease in wild boars and domestic pigs. Lack of effective vaccines hampers the control of virus spread, thus increasing the pressure on the scientific community for urgent solutions. However, knowledge on the immune components associated with protection is very limited. Here we characterized the in vitro recall response induced by immune cells from pigs intranasally vaccinated with the BA71ΔCD2 deletion mutant virus. Vaccination conferred dose-dependent cross-protection associated with both ASFV-specific antibodies and IFNγ-secreting cells. Importantly, bulk and single-cell transcriptomics of blood and lymph node cells from vaccinated pigs revealed a positive feedback from adaptive to innate immunity. Indeed, activation of Th1 and cytotoxic T cells was concomitant with a rapid IFNγ-dependent triggering of an inflammatory response characterized by TNF-producing macrophages, as well as CXCL10-expressing lymphocytes and cross-presenting dendritic cells. Altogether, this study provides a detailed phenotypic characterization of the immune cell subsets involved in cross-protection against ASFV, and highlights key functional immune mechanisms to be considered for the development of an effective ASF vaccine. African swine fever (ASF) pandemic is currently the number one threat for the porcine industry worldwide. Lack of treatments hampers its control, and the insufficient knowledge regarding the immune effector mechanisms required for protection hinders rational vaccine design. Here we present the first comprehensive study characterizing the complex cellular immune response involved in cross-protection against ASF. We show that, upon in vitro reactivation, cells from immune pigs induce a Th1-biased recall response that in turn enhances the antiviral innate response. Our results suggest that this positive feedback regulation of innate immunity plays a key role in the early control of ASF virus infection. Altogether, this work represents a step forward in the understanding of ASF immunology and provide critical immune components that should be considered to more rationally design future ASF vaccines.
Collapse
Affiliation(s)
- Laia Bosch-Camós
- Unitat mixta d’Investigació IRTA-UAB en Sanitat Animal. Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
- IRTA. Programa de Sanitat Animal. Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
| | - Uxía Alonso
- Unitat mixta d’Investigació IRTA-UAB en Sanitat Animal. Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
- IRTA. Programa de Sanitat Animal. Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
| | - Anna Esteve-Codina
- CNAG-CRG, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Chia-Yu Chang
- Unitat mixta d’Investigació IRTA-UAB en Sanitat Animal. Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
- IRTA. Programa de Sanitat Animal. Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
| | - Beatriz Martín-Mur
- CNAG-CRG, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Francesc Accensi
- Unitat mixta d’Investigació IRTA-UAB en Sanitat Animal. Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
- Departament de Sanitat i Anatomia animals. Facultat de Veterinària, Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
| | - Marta Muñoz
- Unitat mixta d’Investigació IRTA-UAB en Sanitat Animal. Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
- IRTA. Programa de Sanitat Animal. Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
| | - María J. Navas
- Unitat mixta d’Investigació IRTA-UAB en Sanitat Animal. Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
- IRTA. Programa de Sanitat Animal. Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
| | - Marc Dabad
- CNAG-CRG, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Enric Vidal
- Unitat mixta d’Investigació IRTA-UAB en Sanitat Animal. Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
- IRTA. Programa de Sanitat Animal. Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
| | - Sonia Pina-Pedrero
- Unitat mixta d’Investigació IRTA-UAB en Sanitat Animal. Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
- IRTA. Programa de Sanitat Animal. Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
| | - Patricia Pleguezuelos
- Unitat mixta d’Investigació IRTA-UAB en Sanitat Animal. Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
- IRTA. Programa de Sanitat Animal. Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
| | - Ginevra Caratù
- CNAG-CRG, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - María L. Salas
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas and Universidad Autònoma de Madrid, Madrid, Spain
| | - Lihong Liu
- National Veterinary Institute (SVA), Uppsala, Sweden
| | | | - Boris Gavrilov
- Biologics Development, Huvepharma, 3A Nikolay Haytov Street, Sofia, Bulgaria
| | - Fernando Rodríguez
- Unitat mixta d’Investigació IRTA-UAB en Sanitat Animal. Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
- IRTA. Programa de Sanitat Animal. Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
- * E-mail: (FR); (JA)
| | - Jordi Argilaguet
- Unitat mixta d’Investigació IRTA-UAB en Sanitat Animal. Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
- IRTA. Programa de Sanitat Animal. Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
- * E-mail: (FR); (JA)
| |
Collapse
|
9
|
Gao Q, Yang Y, Luo Y, Zheng J, Gong L, Wang H, Feng Y, Gong T, Wu D, Wu R, Zheng X, Zheng Z, Zhang G. Adaptation of African swine fever virus to porcine kidney cells stably expressing CD163 and Siglec1. Front Immunol 2022; 13:1015224. [PMID: 36389805 PMCID: PMC9647134 DOI: 10.3389/fimmu.2022.1015224] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 09/27/2022] [Indexed: 08/12/2023] Open
Abstract
African swine fever virus (ASFV) is a complex large DNA enveloped virus that causes African swine fever (ASF) with a fatality rate of up to 100%, seriously threatening the global swine industry. Due to the strict cell tropism of ASFV, there is no effective in vitro cell line, which hinders its prevention and control. Herein, we analyzed genome-wide transcriptional profiles of ASFV-susceptible porcine alveolar macrophages (PAMs) and non-susceptible cell lines PK15 and 3D4-21, an found that PAM surface pattern recognition receptors (PRRs) were significantly higher and common differential genes were significantly enriched in phagocytosis compared with that observed in PK15 and 3D4-21 cell lines. Therefore, endocytosis functions of host cell surface PRRs may play key roles in ASFV infection in vitro. ASFV was found to be infective to PK15 and 3D4-21 cell lines overexpressing CD163 and Siglec1, and to the PK15S1-CD163 cell line stably expressing CD163 and Siglec1. However, the PK15 and 3D4-21 cell lines overexpressing CD163 or Siglec1 alone were not infectious. Simultaneous interference of CD163 and Siglec1 in PAMs with small interfering RNA (siRNA) significantly reduced the infectivity of ASFV. However, siRNA interference of CD163 and Siglec1 respectively did not affect ASFV infectivity. ASFV significantly inhibited IFN expression levels in PAMs and PK15S1-CD163 cells, but had no effect on PK15 and 3D4-21 cell lines. These results indicate that CD163 and Siglec1 are key receptors for ASFV-infected host cells, and both play a synergistic role in the process of ASFV infection. ASFV inhibits IFN expression in susceptible cells, thereby downregulating the host immune response and evading the immune mechanism. The discovery of the ASFV receptor provides novel ideas to study ASFV and host cell interactions, pathogenic mechanisms, development of receptor blockers, vaccine design, and disease resistance breeding.
Collapse
Affiliation(s)
- Qi Gao
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- African Swine Fever Regional Laboratory of China (Guangzhou), Guangzhou, China
- Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Maoming, China
| | - Yunlong Yang
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- African Swine Fever Regional Laboratory of China (Guangzhou), Guangzhou, China
| | - Yizhuo Luo
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- African Swine Fever Regional Laboratory of China (Guangzhou), Guangzhou, China
- Research Center for African Swine Fever Prevention and Control, South China Agricultural University, Guangzhou, China
| | - Jiachen Zheng
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Key Laboratory of Animal Vaccine Development, Ministry of Agriculture and Rural Affairs, Guangzhou, China
| | - Lang Gong
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- African Swine Fever Regional Laboratory of China (Guangzhou), Guangzhou, China
- Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Maoming, China
| | - Heng Wang
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- African Swine Fever Regional Laboratory of China (Guangzhou), Guangzhou, China
- Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Maoming, China
| | - Yongzhi Feng
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Maoming, China
| | - Ting Gong
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Key Laboratory of Animal Vaccine Development, Ministry of Agriculture and Rural Affairs, Guangzhou, China
| | - Dongdong Wu
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Key Laboratory of Animal Vaccine Development, Ministry of Agriculture and Rural Affairs, Guangzhou, China
| | - Ruixia Wu
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Key Laboratory of Animal Vaccine Development, Ministry of Agriculture and Rural Affairs, Guangzhou, China
| | - Xiaoyu Zheng
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Maoming, China
| | - Zezhong Zheng
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- African Swine Fever Regional Laboratory of China (Guangzhou), Guangzhou, China
- Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Maoming, China
- Research Center for African Swine Fever Prevention and Control, South China Agricultural University, Guangzhou, China
| | - Guihong Zhang
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- African Swine Fever Regional Laboratory of China (Guangzhou), Guangzhou, China
- Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Maoming, China
- Research Center for African Swine Fever Prevention and Control, South China Agricultural University, Guangzhou, China
- Key Laboratory of Animal Vaccine Development, Ministry of Agriculture and Rural Affairs, Guangzhou, China
| |
Collapse
|
10
|
Štěpánová H, Kavanová L, Levá L, Vícenová M, Šťastný K, Faldyna M. Characterization of Porcine Monocyte-Derived Macrophages Cultured in Serum-Reduced Medium. BIOLOGY 2022; 11:1457. [PMID: 36290361 PMCID: PMC9598231 DOI: 10.3390/biology11101457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 09/21/2022] [Accepted: 09/27/2022] [Indexed: 11/17/2022]
Abstract
The aim of this study was to establish a cell culture system for the generation of porcine monocyte-derived macrophages (MDMs) under reduced-serum conditions. Cultures based on either the Nu-Serum™ Growth Medium Supplement (NUS) or a conventional fetal bovine serum (FBS) were compared, which included the assessment of FBS from two different providers (FBS1 and FBS2). The data obtained confirmed the significant impact of culture conditions on in vitro-generated MDMs. The MDMs cultured under reduced-serum conditions showed increased levels of IL-1β and CD86 mRNA and a proinflammatory cytokine profile, characterized by the increased mRNA expression of IL-23p19, CXCL10, and CCL5. Phagocytic and respiratory burst activities were not adversely affected. Surprisingly, the difference between the two FBSs was much more pronounced than the effect of the reduced-serum supplement. The FBS1 culture conditions gave rise to macrophages with higher surface levels of CD14, CD16, and CD163, a lower CD80 mRNA expression, and an increased induction of IL-10 gene expression. In contrast, none of these trends were observed in macrophage cultures supplemented with FBS2. Instead, the FBS2 culture showed increased levels of IL-1b and CD86 mRNA. In conclusion, reduced-serum culture is a useful tool for in vitro porcine MDM generation, in line with the current research trend of reducing FBS use in biological research.
Collapse
Affiliation(s)
- Hana Štěpánová
- Veterinary Research Institute, 62100 Brno, Czech Republic
| | | | | | | | | | | |
Collapse
|
11
|
Kwon HI, Do DT, Van Vo H, Lee SC, Kim MH, Nguyen DTT, Tran TM, Le QTV, Ngo TTN, Nguyen NM, Lee JY, Nguyen TT. Development of optimized protocol for culturing African swine fever virus field isolates in MA104 cells. CANADIAN JOURNAL OF VETERINARY RESEARCH = REVUE CANADIENNE DE RECHERCHE VETERINAIRE 2022; 86:261-268. [PMID: 36211218 PMCID: PMC9536354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 03/23/2022] [Indexed: 04/03/2023]
Abstract
The goal of this study was to identify a candidate commercial cell line for the replication of African swine fever virus (ASFV) by comparing several available cell lines with various medium factors. In the sensitivity test of cells, MA104 and MARC-145 had strong potential for ASFV replication. Next, MA104 cells were used to compare the adaptation of ASFV obtained from tissue homogenates and blood samples in various infectious media. At the 10th passage, the ASFV obtained from the blood sample had a significantly higher viral load than that obtained from the tissue sample (P = 0.000), exhibiting a mean cycle threshold (Ct) value = 20.39 ± 1.99 compared with 25.36 ± 2.11. For blood samples, ASFV grew on infectious medium B more robustly than on infectious medium A (P = 0.006), corresponding to a Ct value = 19.58 ± 2.10 versus 21.20 ± 1.47. African swine fever virus originating from blood specimens continued to multiply gradually and peaked in the 15th passage, exhibiting a Ct value = 14.36 ± 0.22 in infectious medium B and a Ct value = 15.42 ± 0.14 in infectious medium A. When ASFV was cultured from tissue homogenates, however, there was no difference (P = 0.062) in ASFV growth between infectious media A and B. A model was developed to enhance ASFV replication through adaptation to MA104 cells. The lack of mutation at the genetic segments encoding p72, p54, p30, and the central hypervariable region (CVR) in serial culture passages is important in increasing the probability of maintaining immunogenicity when developing a vaccine candidate.
Collapse
Affiliation(s)
- Hyeok-Il Kwon
- ChoongAng Vaccine Laboratories, Daejeon, 34055, Republic of Korea (Hyeok-il Kwon, Seung-Chul Lee, Min Ho Kim, Joo Young Lee); Department of Infectious Diseases and Veterinary Public Health, Faculty of Animal Science and Veterinary Medicine, Nong Lam University, Thu Duc District, Ho Chi Minh City, Vietnam (Duy Tien Do, Dung Thi Thuy Nguyen, Tram Thi Ngoc Ngo, Toan Tat Nguyen); Center for Veterinary Diagnostics, Regional Animal Health Office No. 6, Ho Chi Minh City, Department of Animal Health, Vietnam (Hung Van Vo, Tan Minh Tran, Quang Tin Vinh Le); Research Center for Genetics and Reproductive Health (CGRH), School of Medicine, National University HCMC, Vietnam, (Nam Minh Nguyen)
| | - Duy Tien Do
- ChoongAng Vaccine Laboratories, Daejeon, 34055, Republic of Korea (Hyeok-il Kwon, Seung-Chul Lee, Min Ho Kim, Joo Young Lee); Department of Infectious Diseases and Veterinary Public Health, Faculty of Animal Science and Veterinary Medicine, Nong Lam University, Thu Duc District, Ho Chi Minh City, Vietnam (Duy Tien Do, Dung Thi Thuy Nguyen, Tram Thi Ngoc Ngo, Toan Tat Nguyen); Center for Veterinary Diagnostics, Regional Animal Health Office No. 6, Ho Chi Minh City, Department of Animal Health, Vietnam (Hung Van Vo, Tan Minh Tran, Quang Tin Vinh Le); Research Center for Genetics and Reproductive Health (CGRH), School of Medicine, National University HCMC, Vietnam, (Nam Minh Nguyen)
| | - Hung Van Vo
- ChoongAng Vaccine Laboratories, Daejeon, 34055, Republic of Korea (Hyeok-il Kwon, Seung-Chul Lee, Min Ho Kim, Joo Young Lee); Department of Infectious Diseases and Veterinary Public Health, Faculty of Animal Science and Veterinary Medicine, Nong Lam University, Thu Duc District, Ho Chi Minh City, Vietnam (Duy Tien Do, Dung Thi Thuy Nguyen, Tram Thi Ngoc Ngo, Toan Tat Nguyen); Center for Veterinary Diagnostics, Regional Animal Health Office No. 6, Ho Chi Minh City, Department of Animal Health, Vietnam (Hung Van Vo, Tan Minh Tran, Quang Tin Vinh Le); Research Center for Genetics and Reproductive Health (CGRH), School of Medicine, National University HCMC, Vietnam, (Nam Minh Nguyen)
| | - Seung-Chul Lee
- ChoongAng Vaccine Laboratories, Daejeon, 34055, Republic of Korea (Hyeok-il Kwon, Seung-Chul Lee, Min Ho Kim, Joo Young Lee); Department of Infectious Diseases and Veterinary Public Health, Faculty of Animal Science and Veterinary Medicine, Nong Lam University, Thu Duc District, Ho Chi Minh City, Vietnam (Duy Tien Do, Dung Thi Thuy Nguyen, Tram Thi Ngoc Ngo, Toan Tat Nguyen); Center for Veterinary Diagnostics, Regional Animal Health Office No. 6, Ho Chi Minh City, Department of Animal Health, Vietnam (Hung Van Vo, Tan Minh Tran, Quang Tin Vinh Le); Research Center for Genetics and Reproductive Health (CGRH), School of Medicine, National University HCMC, Vietnam, (Nam Minh Nguyen)
| | - Min Ho Kim
- ChoongAng Vaccine Laboratories, Daejeon, 34055, Republic of Korea (Hyeok-il Kwon, Seung-Chul Lee, Min Ho Kim, Joo Young Lee); Department of Infectious Diseases and Veterinary Public Health, Faculty of Animal Science and Veterinary Medicine, Nong Lam University, Thu Duc District, Ho Chi Minh City, Vietnam (Duy Tien Do, Dung Thi Thuy Nguyen, Tram Thi Ngoc Ngo, Toan Tat Nguyen); Center for Veterinary Diagnostics, Regional Animal Health Office No. 6, Ho Chi Minh City, Department of Animal Health, Vietnam (Hung Van Vo, Tan Minh Tran, Quang Tin Vinh Le); Research Center for Genetics and Reproductive Health (CGRH), School of Medicine, National University HCMC, Vietnam, (Nam Minh Nguyen)
| | - Dung Thi Thuy Nguyen
- ChoongAng Vaccine Laboratories, Daejeon, 34055, Republic of Korea (Hyeok-il Kwon, Seung-Chul Lee, Min Ho Kim, Joo Young Lee); Department of Infectious Diseases and Veterinary Public Health, Faculty of Animal Science and Veterinary Medicine, Nong Lam University, Thu Duc District, Ho Chi Minh City, Vietnam (Duy Tien Do, Dung Thi Thuy Nguyen, Tram Thi Ngoc Ngo, Toan Tat Nguyen); Center for Veterinary Diagnostics, Regional Animal Health Office No. 6, Ho Chi Minh City, Department of Animal Health, Vietnam (Hung Van Vo, Tan Minh Tran, Quang Tin Vinh Le); Research Center for Genetics and Reproductive Health (CGRH), School of Medicine, National University HCMC, Vietnam, (Nam Minh Nguyen)
| | - Tan Minh Tran
- ChoongAng Vaccine Laboratories, Daejeon, 34055, Republic of Korea (Hyeok-il Kwon, Seung-Chul Lee, Min Ho Kim, Joo Young Lee); Department of Infectious Diseases and Veterinary Public Health, Faculty of Animal Science and Veterinary Medicine, Nong Lam University, Thu Duc District, Ho Chi Minh City, Vietnam (Duy Tien Do, Dung Thi Thuy Nguyen, Tram Thi Ngoc Ngo, Toan Tat Nguyen); Center for Veterinary Diagnostics, Regional Animal Health Office No. 6, Ho Chi Minh City, Department of Animal Health, Vietnam (Hung Van Vo, Tan Minh Tran, Quang Tin Vinh Le); Research Center for Genetics and Reproductive Health (CGRH), School of Medicine, National University HCMC, Vietnam, (Nam Minh Nguyen)
| | - Quang Tin Vinh Le
- ChoongAng Vaccine Laboratories, Daejeon, 34055, Republic of Korea (Hyeok-il Kwon, Seung-Chul Lee, Min Ho Kim, Joo Young Lee); Department of Infectious Diseases and Veterinary Public Health, Faculty of Animal Science and Veterinary Medicine, Nong Lam University, Thu Duc District, Ho Chi Minh City, Vietnam (Duy Tien Do, Dung Thi Thuy Nguyen, Tram Thi Ngoc Ngo, Toan Tat Nguyen); Center for Veterinary Diagnostics, Regional Animal Health Office No. 6, Ho Chi Minh City, Department of Animal Health, Vietnam (Hung Van Vo, Tan Minh Tran, Quang Tin Vinh Le); Research Center for Genetics and Reproductive Health (CGRH), School of Medicine, National University HCMC, Vietnam, (Nam Minh Nguyen)
| | - Tram Thi Ngoc Ngo
- ChoongAng Vaccine Laboratories, Daejeon, 34055, Republic of Korea (Hyeok-il Kwon, Seung-Chul Lee, Min Ho Kim, Joo Young Lee); Department of Infectious Diseases and Veterinary Public Health, Faculty of Animal Science and Veterinary Medicine, Nong Lam University, Thu Duc District, Ho Chi Minh City, Vietnam (Duy Tien Do, Dung Thi Thuy Nguyen, Tram Thi Ngoc Ngo, Toan Tat Nguyen); Center for Veterinary Diagnostics, Regional Animal Health Office No. 6, Ho Chi Minh City, Department of Animal Health, Vietnam (Hung Van Vo, Tan Minh Tran, Quang Tin Vinh Le); Research Center for Genetics and Reproductive Health (CGRH), School of Medicine, National University HCMC, Vietnam, (Nam Minh Nguyen)
| | - Nam Minh Nguyen
- ChoongAng Vaccine Laboratories, Daejeon, 34055, Republic of Korea (Hyeok-il Kwon, Seung-Chul Lee, Min Ho Kim, Joo Young Lee); Department of Infectious Diseases and Veterinary Public Health, Faculty of Animal Science and Veterinary Medicine, Nong Lam University, Thu Duc District, Ho Chi Minh City, Vietnam (Duy Tien Do, Dung Thi Thuy Nguyen, Tram Thi Ngoc Ngo, Toan Tat Nguyen); Center for Veterinary Diagnostics, Regional Animal Health Office No. 6, Ho Chi Minh City, Department of Animal Health, Vietnam (Hung Van Vo, Tan Minh Tran, Quang Tin Vinh Le); Research Center for Genetics and Reproductive Health (CGRH), School of Medicine, National University HCMC, Vietnam, (Nam Minh Nguyen)
| | - Joo Young Lee
- ChoongAng Vaccine Laboratories, Daejeon, 34055, Republic of Korea (Hyeok-il Kwon, Seung-Chul Lee, Min Ho Kim, Joo Young Lee); Department of Infectious Diseases and Veterinary Public Health, Faculty of Animal Science and Veterinary Medicine, Nong Lam University, Thu Duc District, Ho Chi Minh City, Vietnam (Duy Tien Do, Dung Thi Thuy Nguyen, Tram Thi Ngoc Ngo, Toan Tat Nguyen); Center for Veterinary Diagnostics, Regional Animal Health Office No. 6, Ho Chi Minh City, Department of Animal Health, Vietnam (Hung Van Vo, Tan Minh Tran, Quang Tin Vinh Le); Research Center for Genetics and Reproductive Health (CGRH), School of Medicine, National University HCMC, Vietnam, (Nam Minh Nguyen)
| | - Toan Tat Nguyen
- ChoongAng Vaccine Laboratories, Daejeon, 34055, Republic of Korea (Hyeok-il Kwon, Seung-Chul Lee, Min Ho Kim, Joo Young Lee); Department of Infectious Diseases and Veterinary Public Health, Faculty of Animal Science and Veterinary Medicine, Nong Lam University, Thu Duc District, Ho Chi Minh City, Vietnam (Duy Tien Do, Dung Thi Thuy Nguyen, Tram Thi Ngoc Ngo, Toan Tat Nguyen); Center for Veterinary Diagnostics, Regional Animal Health Office No. 6, Ho Chi Minh City, Department of Animal Health, Vietnam (Hung Van Vo, Tan Minh Tran, Quang Tin Vinh Le); Research Center for Genetics and Reproductive Health (CGRH), School of Medicine, National University HCMC, Vietnam, (Nam Minh Nguyen)
| |
Collapse
|
12
|
African Swine Fever Vaccinology: The Biological Challenges from Immunological Perspectives. Viruses 2022; 14:v14092021. [PMID: 36146827 PMCID: PMC9505361 DOI: 10.3390/v14092021] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 08/22/2022] [Accepted: 09/08/2022] [Indexed: 11/17/2022] Open
Abstract
African swine fever virus (ASFV), a nucleocytoplasmic large DNA virus (NCLDV), causes African swine fever (ASF), an acute hemorrhagic disease with mortality rates up to 100% in domestic pigs. ASF is currently epidemic or endemic in many countries and threatening the global swine industry. Extensive ASF vaccine research has been conducted since the 1920s. Like inactivated viruses of other NCLDVs, such as vaccinia virus, inactivated ASFV vaccine candidates did not induce protective immunity. However, inactivated lumpy skin disease virus (poxvirus) vaccines are protective in cattle. Unlike some experimental poxvirus subunit vaccines that induced protection, ASF subunit vaccine candidates implemented with various platforms containing several ASFV structural genes or proteins failed to protect pigs effectively. Only some live attenuated viruses (LAVs) are able to protect pigs with high degrees of efficacy. There are currently several LAV ASF vaccine candidates. Only one commercial LAV vaccine is approved for use in Vietnam. LAVs, as ASF vaccines, have not yet been widely tested. Reports thus far show that the onset and duration of protection induced by the LAVs are late and short, respectively, compared to LAV vaccines for other diseases. In this review, the biological challenges in the development of ASF vaccines, especially subunit platforms, are discussed from immunological perspectives based on several unusual ASFV characteristics shared with HIV and poxviruses. These characteristics, including multiple distinct infectious virions, extremely high glycosylation and low antigen surface density of envelope proteins, immune evasion, and possible apoptotic mimicry, could pose enormous challenges to the development of ASF vaccines, especially subunit platforms designed to induce humoral immunity.
Collapse
|
13
|
Jarosova R, Ondrackova P, Leva L, Nedbalcova K, Vicenova M, Masek J, Volf J, Gebauer J, Do T, Guran R, Sladek Z, Dominguez J, Faldyna M. Cytokine expression by CD163+ monocytes in healthy and Actinobacillus pleuropneumoniae-infected pigs. Res Vet Sci 2022; 152:1-9. [PMID: 35901636 DOI: 10.1016/j.rvsc.2022.07.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Revised: 06/30/2022] [Accepted: 07/18/2022] [Indexed: 11/29/2022]
Abstract
Distinct monocyte subpopulations have been previously described in healthy pigs and pigs experimentally infected with Actinobacillus pleuropneumoniae (APP). The CD163+ subpopulation of bone marrow (BM), peripheral blood (PB) and lung monocytes was found to play an important role in the inflammatory process. The inflammation is accompanied by elevation of inflammatory cytokines. The aim of the study was to evaluate the contribution of CD163+ monocytes and macrophages to cytokine production during APP-induced lung inflammation. Cytokine production was assessed by flow cytometry (FC) and quantitative PCR (qPCR) in CD163+ monocytes and by qPCR, immunohistochemistry/fluorescence in lungs and tracheobronchial lymph nodes (TBLN). Despite the systemic inflammatory response after APP infection, BM and PB CD163+ monocytes did not express elevated levels of a wide range of cytokines compared to control pigs. In contrast, significant amounts of IL-1β, IL-6, IL-8 and TNF-α were produced in lung lesions and IL-1β in the TBLN. At the protein level, TNF-α was expressed by both CD163+ monocytes and macrophages in lung lesions, whereas IL-1β, IL-6 and IL-8 expression was found only in CD163+ monocytes; no CD163+ macrophages were found to produce these cytokines. Furthermore, the quantification of CD163+ monocytes expressing the two cytokines IL-1β and IL-8 that were most elevated was performed. In lung lesions, 36.5% IL-1β positive CD163+ monocytes but only 18.3% IL-8 positive CD163+ monocytes were found. In conclusion, PB and BM CD163+ monocytes do not appear to contribute to the elevated cytokine levels in plasma. On the other hand, CD163+ monocytes contribute to inflammatory cytokine expression, especially IL-1β at the site of inflammation during the inflammatory process.
Collapse
Affiliation(s)
- Rea Jarosova
- Veterinary Research Institute, Brno, Czech Republic; Department of Morphology, Physiology and Animal Genetics, The Faculty of AgriSciences, Mendel University in Brno, Brno, Czech Republic.
| | | | - Lenka Leva
- Veterinary Research Institute, Brno, Czech Republic.
| | | | | | - Josef Masek
- Veterinary Research Institute, Brno, Czech Republic.
| | - Jiri Volf
- Veterinary Research Institute, Brno, Czech Republic.
| | - Jan Gebauer
- Veterinary Research Institute, Brno, Czech Republic.
| | - Tomas Do
- Department of Chemistry and Biochemistry, Faculty of AgriSciences, Mendel University in Brno, Brno, Czech Republic.
| | - Roman Guran
- Department of Chemistry and Biochemistry, Faculty of AgriSciences, Mendel University in Brno, Brno, Czech Republic; Central European Institute of Technology, Brno University of Technology, Brno, Czech Republic
| | - Zbysek Sladek
- Department of Morphology, Physiology and Animal Genetics, The Faculty of AgriSciences, Mendel University in Brno, Brno, Czech Republic.
| | - Javier Dominguez
- Departmento de Biotecnologia, Centro Nacional Instituto de Investigacion y Tecnologia Agraria y Alimentaria (CSIC-INIA), Madrid, Spain.
| | | |
Collapse
|
14
|
Canter JA, Aponte T, Ramirez-Medina E, Pruitt S, Gladue DP, Borca MV, Zhu JJ. Serum Neutralizing and Enhancing Effects on African Swine Fever Virus Infectivity in Adherent Pig PBMC. Viruses 2022; 14:v14061249. [PMID: 35746720 PMCID: PMC9229155 DOI: 10.3390/v14061249] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 06/03/2022] [Accepted: 06/07/2022] [Indexed: 12/28/2022] Open
Abstract
African swine fever virus (ASFV) causes hemorrhagic fever with mortality rates of up to 100% in domestic pigs. Currently, there are no commercial vaccines for the disease. Only some live-attenuated viruses have been able to protect pigs from ASFV infection. The immune mechanisms involved in the protection are unclear. Immune sera can neutralize ASFV but incompletely. The mechanisms involved are not fully understood. Currently, there is no standardized protocol for ASFV neutralization assays. In this study, a flow cytometry-based ASFV neutralization assay was developed and tested in pig adherent PBMC using a virulent ASFV containing a fluorescent protein gene as a substrate for neutralization. As with previous studies, the percentage of infected macrophages was approximately five time higher than that of infected monocytes, and nearly all infected cells displayed no staining with anti-CD16 antibodies. Sera from naïve pigs and pigs immunized with a live-attenuated ASFV and fully protected against parental virus were used in the assay. The sera displayed incomplete neutralization with MOI-dependent neutralizing efficacies. Extracellular, but not intracellular, virions suspended in naïve serum were more infectious than those in the culture medium, as reported for some enveloped viruses, suggesting a novel mechanism of ASFV infection in macrophages. Both the intracellular and extracellular virions could not be completely neutralized.
Collapse
Affiliation(s)
- Jessica A. Canter
- Foreign Animal Disease Research Unit, Plum Island Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Orient, NY 11957, USA; (J.A.C.); (T.A.); (E.R.-M.); (S.P.)
- Plum Island Animal Disease Center, Oak Ridge Institute for Science and Education, Oak Ridge, TN 37830, USA
| | - Theresa Aponte
- Foreign Animal Disease Research Unit, Plum Island Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Orient, NY 11957, USA; (J.A.C.); (T.A.); (E.R.-M.); (S.P.)
- Plum Island Animal Disease Center, Oak Ridge Institute for Science and Education, Oak Ridge, TN 37830, USA
| | - Elizabeth Ramirez-Medina
- Foreign Animal Disease Research Unit, Plum Island Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Orient, NY 11957, USA; (J.A.C.); (T.A.); (E.R.-M.); (S.P.)
| | - Sarah Pruitt
- Foreign Animal Disease Research Unit, Plum Island Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Orient, NY 11957, USA; (J.A.C.); (T.A.); (E.R.-M.); (S.P.)
| | - Douglas P. Gladue
- Foreign Animal Disease Research Unit, Plum Island Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Orient, NY 11957, USA; (J.A.C.); (T.A.); (E.R.-M.); (S.P.)
- Correspondence: (D.P.G.); (M.V.B.); (J.J.Z.); Tel.: +1-631-323-3131 (D.P.G.); +1-631-323-3035 (M.V.B.); +1-631-323-3186 (J.J.Z.)
| | - Manuel V. Borca
- Foreign Animal Disease Research Unit, Plum Island Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Orient, NY 11957, USA; (J.A.C.); (T.A.); (E.R.-M.); (S.P.)
- Correspondence: (D.P.G.); (M.V.B.); (J.J.Z.); Tel.: +1-631-323-3131 (D.P.G.); +1-631-323-3035 (M.V.B.); +1-631-323-3186 (J.J.Z.)
| | - James J. Zhu
- Foreign Animal Disease Research Unit, Plum Island Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Orient, NY 11957, USA; (J.A.C.); (T.A.); (E.R.-M.); (S.P.)
- Correspondence: (D.P.G.); (M.V.B.); (J.J.Z.); Tel.: +1-631-323-3131 (D.P.G.); +1-631-323-3035 (M.V.B.); +1-631-323-3186 (J.J.Z.)
| |
Collapse
|
15
|
Meloni D, Franzoni G, Oggiano A. Cell Lines for the Development of African Swine Fever Virus Vaccine Candidates: An Update. Vaccines (Basel) 2022; 10:707. [PMID: 35632463 PMCID: PMC9144233 DOI: 10.3390/vaccines10050707] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 04/27/2022] [Accepted: 04/27/2022] [Indexed: 01/27/2023] Open
Abstract
African swine fever virus (ASFV) is the etiological agent of a highly lethal disease in both domestic and wild pigs. The virus has rapidly spread worldwide and has no available licensed vaccine. An obstacle to the construction of a safe and efficient vaccine is the lack of a suitable cell line for ASFV isolation and propagation. Macrophages are the main targets for ASFV, and they have been widely used to study virus-host interactions; nevertheless, obtaining these cells is time-consuming and expensive, and they are not ethically suitable for the production of large-scale vaccines. To overcome these issues, different virulent field isolates have been adapted on monkey or human continuous cells lines; however, several culture passages often lead to significant genetic modifications and the loss of immunogenicity of the adapted strain. Thus, several groups have attempted to establish a porcine cell line able to sustain ASFV growth. Preliminary data suggested that some porcine continuous cell lines might be an alternative to primary macrophages for ASFV research and for large-scale vaccine production, although further studies are still needed. In this review, we summarize the research to investigate the most suitable cell line for ASFV isolation and propagation.
Collapse
Affiliation(s)
| | - Giulia Franzoni
- Department of Animal Health, Istituto Zooprofilattico Sperimentale della Sardegna, 07100 Sassari, Italy; (D.M.); (A.O.)
| | | |
Collapse
|
16
|
Lin SF, Lin HA, Chuang HC, Tsai HW, Kuo N, Chen SC, Hou SK. Fever, Tachypnea, and Monocyte Distribution Width Predicts Length of Stay for Patients with COVID-19: A Pioneer Study. J Pers Med 2022; 12:jpm12030449. [PMID: 35330449 PMCID: PMC8953796 DOI: 10.3390/jpm12030449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/04/2022] [Accepted: 03/11/2022] [Indexed: 12/04/2022] Open
Abstract
(1) Background: Our study investigated whether monocyte distribution width (MDW) could be used in emergency department (ED) settings as a predictor of prolonged length of stay (LOS) for patients with COVID-19. (2) Methods: A retrospective cohort study was conducted; patients presenting to the ED of an academic hospital with confirmed COVID-19 were enrolled. Multivariable logistic regression models were used to obtain the odds ratios (ORs) for predictors of an LOS of >14 days. A validation study for the association between MDW and cycle of threshold (Ct) value was performed. (3) Results: Fever > 38 °C (OR: 2.82, 95% CI, 1.13−7.02, p = 0.0259), tachypnea (OR: 4.76, 95% CI, 1.67−13.55, p = 0.0034), and MDW ≥ 21 (OR: 5.67, 95% CI, 1.19−27.10, p = 0.0269) were robust significant predictors of an LOS of >14 days. We developed a new scoring system in which patients were assigned 1 point for fever > 38 °C, 2 points for tachypnea > 20 breath/min, and 3 points for MDW ≥ 21. The optimal cutoff was a score of ≥2. MDW was negatively associated with Ct value (β: −0.32 per day, standard error = 0.12, p = 0.0099). (4) Conclusions: Elevated MDW was associated with a prolonged LOS.
Collapse
Affiliation(s)
- Sheng-Feng Lin
- Department of Public Health, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan;
- School of Public Health, College of Public Health, Taipei Medical University, Taipei 110, Taiwan
- Department of Emergency Medicine, Taipei Medical University Hospital, Taipei 110, Taiwan; (H.-A.L.); (H.-W.T.); (N.K.); (S.-C.C.)
| | - Hui-An Lin
- Department of Emergency Medicine, Taipei Medical University Hospital, Taipei 110, Taiwan; (H.-A.L.); (H.-W.T.); (N.K.); (S.-C.C.)
- Graduate Institute of Injury Prevention and Control, College of Public Health, Taipei Medical University, Taipei 110, Taiwan
| | - Han-Chuan Chuang
- Division of Infectious Diseases, Department of Internal Medicine, Taipei Medical University Hospital, Taipei 110, Taiwan;
| | - Hung-Wei Tsai
- Department of Emergency Medicine, Taipei Medical University Hospital, Taipei 110, Taiwan; (H.-A.L.); (H.-W.T.); (N.K.); (S.-C.C.)
| | - Ning Kuo
- Department of Emergency Medicine, Taipei Medical University Hospital, Taipei 110, Taiwan; (H.-A.L.); (H.-W.T.); (N.K.); (S.-C.C.)
| | - Shao-Chun Chen
- Department of Emergency Medicine, Taipei Medical University Hospital, Taipei 110, Taiwan; (H.-A.L.); (H.-W.T.); (N.K.); (S.-C.C.)
| | - Sen-Kuang Hou
- Department of Emergency Medicine, Taipei Medical University Hospital, Taipei 110, Taiwan; (H.-A.L.); (H.-W.T.); (N.K.); (S.-C.C.)
- Department of Emergency Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
- Correspondence: ; Tel.: +886-2-2736-1661 (ext. 8107)
| |
Collapse
|
17
|
Zimmermann CE, Mackens-Kiani L, Acil Y, Terheyden H. Characterization of porcine mesenchymal stromal cells and their proliferative and osteogenic potential in long-term culture. J Stem Cells Regen Med 2022; 17:49-55. [PMID: 35250201 DOI: 10.46582/jsrm.1702008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 10/07/2021] [Indexed: 12/29/2022]
Abstract
Background: Porcine mesenchymal stromal cells (pMSCs) are considered a valuable research model for bone tissue engineering, which requires adequate amounts of viable cells with sufficient potential for osteogenic differentiation. For isolation and expansion of these cells through long-term culture, appropriate culture conditions are needed. Objective: To study the effect of extended in vitro cultivation on pMSC proliferation and differentiation potential using different osteogenic and adipogenic induction media. Methods: pMSCs were isolated from the bone marrow of adult Göttingen minipigs, cultured, expanded to passage 20 (~160 days) and characterized by their expression of cell surface markers (wCD44, CD45, CD90, SWC9, fibronectin), alkaline phosphatase (ALP), and osteocalcin and their potential for osteogenic and adipogenic differentiation using different induction media. Results: pMSCs retained their capacity for proliferation and osteogenic differentiation, and the number of CD90-positive cells increased significantly over more than 60 population doublings. CD90 expression in uninduced cells correlated strongly with ALP expression following osteogenic induction. Medium enriched with calcium yielded a stronger osteogenic response. Conclusion: The selection of CD90-positive MSCs and adequate levels of calcium seem to enhance the osteogenic phenotype for bone tissue engineering.
Collapse
Affiliation(s)
- Corinna E Zimmermann
- Department of Craniomaxillofacial Surgery, University Hospital Schleswig-Holstein, Campus Kiel, Arnold-Heller-Strasse 3, 24105 Kiel, Germany.,University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany
| | | | - Yahya Acil
- Department of Craniomaxillofacial Surgery, University Hospital Schleswig-Holstein, Campus Kiel, Arnold-Heller-Strasse 3, 24105 Kiel, Germany
| | - Hendrik Terheyden
- Department of Craniomaxillofacial Surgery, University Hospital Schleswig-Holstein, Campus Kiel, Arnold-Heller-Strasse 3, 24105 Kiel, Germany
| |
Collapse
|
18
|
A Multi-Laboratory Comparison of Methods for Detection and Quantification of African Swine Fever Virus. Pathogens 2022; 11:pathogens11030325. [PMID: 35335649 PMCID: PMC8949307 DOI: 10.3390/pathogens11030325] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/26/2022] [Accepted: 03/03/2022] [Indexed: 02/01/2023] Open
Abstract
African swine fever is a viral disease of the family Suidae. Methods to detect and quantify African swine fever virus (ASFV) include qPCR and virus infectivity assays. Individual laboratories often use in-house procedures for these assays, which can hamper the comparison of results. The objective of this study was to estimate the probability of ASFV detection using these assays, and to determine the inter-test correlations between results. This was achieved by testing a panel of 80 samples at three reference laboratories. Samples were analysed using nucleic acid extraction and qPCR, as well as virus infectivity assays. For qPCR, a very high probability (ranging from 0.96 to 1.0) of detecting ASFV DNA was observed for all tested systems. For virus infectivity assays in cells, the probability of detecting infectious ASFV varied from 0.68 to 0.90 and was highest using pulmonary alveolar macrophages, followed by MARC145 cells, peripheral blood monocytes, and finally wild boar lung cells. Intraclass correlation coefficient estimates of 0.97 (0.96–0.98) between qPCR methods, 0.80 (0.74–0.85) to 0.94 (0.92–0.96) between virus infectivity assays, and 0.77 (0.68–0.83) to 0.95 (0.93–0.96) between qPCR methods and virus infectivity assays were obtained. These findings show that qPCR gives the highest probability for the detection of ASFV.
Collapse
|
19
|
Banete A, Barilo J, Whittaker R, Basta S. The Activated Macrophage - A Tough Fortress for Virus Invasion: How Viruses Strike Back. Front Microbiol 2022; 12:803427. [PMID: 35087503 PMCID: PMC8787342 DOI: 10.3389/fmicb.2021.803427] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 12/06/2021] [Indexed: 12/29/2022] Open
Abstract
Macrophages (Mφ) are innate immune cells with a variety of functional phenotypes depending on the cytokine microenvironment they reside in. Mφ exhibit distinct activation patterns that are found within a wide array of activation states ranging from the originally discovered classical pro-inflammatory (M1) to the anti-inflammatory (M2) with their multi-facades. M1 cells are induced by IFNγ + LPS, while M2 are further subdivided into M2a (IL-4), M2b (Immune Complex) and M2c (IL-10) based on their inducing stimuli. Not surprisingly, Mφ activation influences the outcome of viral infections as they produce cytokines that in turn activate cells of the adaptive immune system. Generally, activated M1 cells tend to restrict viral replication, however, influenza and HIV exploit inflammation to support their replication. Moreover, M2a polarization inhibits HIV replication at the post-integration level, while HCMV encoded hrIL-10 suppresses inflammatory reactions by facilitating M2c formation. Additionally, viruses such as LCMV and Lassa Virus directly suppress Mφ activation leading to viral chronicity. Here we review how Mφ activation affects viral infection and the strategies by which viruses manipulate Mφ polarization to benefit their own fitness. An understanding of these mechanisms is important for the development of novel immunotherapies that can sway Mφ phenotype to inhibit viral replication.
Collapse
Affiliation(s)
- Andra Banete
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada.,Department of Biological Sciences, Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada
| | - Julia Barilo
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| | - Reese Whittaker
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| | - Sameh Basta
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| |
Collapse
|
20
|
Razzuoli E, Armando F, De Paolis L, Ciurkiewicz M, Amadori M. The Swine IFN System in Viral Infections: Major Advances and Translational Prospects. Pathogens 2022; 11:175. [PMID: 35215119 PMCID: PMC8875149 DOI: 10.3390/pathogens11020175] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/22/2022] [Accepted: 01/24/2022] [Indexed: 02/01/2023] Open
Abstract
Interferons (IFNs) are a family of cytokines that play a pivotal role in orchestrating the innate immune response during viral infections, thus representing the first line of defense in the host. After binding to their respective receptors, they are able to elicit a plethora of biological activities, by initiating signaling cascades which lead to the transcription of genes involved in antiviral, anti-inflammatory, immunomodulatory and antitumoral effector mechanisms. In hindsight, it is not surprising that viruses have evolved multiple IFN escape strategies toward efficient replication in the host. Hence, in order to achieve insight into preventive and treatment strategies, it is essential to explore the mechanisms underlying the IFN response to viral infections and the constraints thereof. Accordingly, this review is focused on three RNA and three DNA viruses of major importance in the swine farming sector, aiming to provide essential data as to how the IFN system modulates the antiviral immune response, and is affected by diverse, virus-driven, immune escape mechanisms.
Collapse
Affiliation(s)
- Elisabetta Razzuoli
- National Reference Center of Veterinary and Comparative Oncology (CEROVEC), Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle D’Aosta, Piazza Borgo Pila 39/24, 16129 Genoa, Italy;
| | - Federico Armando
- Department of Pathology, University of Veterinary Medicine Hannover, Bünteweg 17, 30559 Hannover, Germany; (F.A.); (M.C.)
| | - Livia De Paolis
- National Reference Center of Veterinary and Comparative Oncology (CEROVEC), Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle D’Aosta, Piazza Borgo Pila 39/24, 16129 Genoa, Italy;
| | - Malgorzata Ciurkiewicz
- Department of Pathology, University of Veterinary Medicine Hannover, Bünteweg 17, 30559 Hannover, Germany; (F.A.); (M.C.)
| | - Massimo Amadori
- National Network of Veterinary Immunology (RNIV), Via Istria 3, 25125 Brescia, Italy;
| |
Collapse
|
21
|
Oh T, Do DT, Lai DC, Nguyen TC, Vo HV, Chae C. Age-related viral load and severity of systemic pathological lesions in acute naturally occurring African swine fever virus genotype II infections. Comp Immunol Microbiol Infect Dis 2021; 79:101709. [PMID: 34543808 DOI: 10.1016/j.cimid.2021.101709] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 08/31/2021] [Accepted: 09/07/2021] [Indexed: 11/17/2022]
Abstract
African swine fever (ASF) causes a contagious hemorrhagic disease in all ages of pigs without sex predilections. The objective of this study was to determine the age-related viral loads and severity of systemic pathological lesions among three different swine group ages (weaned pigs, fattening pigs, and sows) during a recent outbreak of acute ASF in Vietnam. Age-related viral loads were determined in 5 major organs (lung, liver, spleen, kidney, and lymph node) by immunohistochemistry as well as in the blood by real-time polymerase chain reaction (PCR). Age-related systemic pathological lesions were analyzed in the listed organs among three age groups. Weaned pigs had significantly (p < 0.05) higher levels of viral loads in their lung, liver, lymph nodes and blood than in those of fattening pigs and sows. Fattening pigs had significantly (p < 0.05) higher scores of macroscopic lung and lymphoid lesions, and microscopic liver lesions compared with those of weaned pigs and sows. The results of this study demonstrated that viral loads were age-related in acute naturally occurring ASF but the severity of pathological lesions was not correlated with the level of viral loads in the five major organs.
Collapse
Affiliation(s)
- Taehwan Oh
- Department of Veterinary Pathology, College of Veterinary Medicine, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea
| | - Duy Tien Do
- Faculty of Animal Sciences and Veterinary Medicine, Nong Lam University, Thu Duc District, Ho Chi Minh City, Viet Nam
| | - Danh Cong Lai
- Faculty of Animal Sciences and Veterinary Medicine, Nong Lam University, Thu Duc District, Ho Chi Minh City, Viet Nam
| | - Thanh Che Nguyen
- Faculty of Animal Sciences and Veterinary Medicine, Nong Lam University, Thu Duc District, Ho Chi Minh City, Viet Nam
| | - Hung Van Vo
- Department of Animal Health, Center for Veterinary Diagnostics, Regional Animal Health Office No. 6, Ho Chi Minh City, Viet Nam
| | - Chanhee Chae
- Department of Veterinary Pathology, College of Veterinary Medicine, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea.
| |
Collapse
|
22
|
Wang Y, Xu Z, Zhou Y, Xie M, Qi X, Xu Z, Cai Q, Sheng H, Chen E, Zhao B, Mao E. Leukocyte cell population data from the blood cell analyzer as a predictive marker for severity of acute pancreatitis. J Clin Lab Anal 2021; 35:e23863. [PMID: 34062621 PMCID: PMC8274994 DOI: 10.1002/jcla.23863] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 05/11/2021] [Accepted: 05/16/2021] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND The prediction for severe acute pancreatitis (SAP) is the key to give timely targeted treatment. Leukocyte cell population data (CPD) have been widely applied in early prediction and diagnosis of many diseases, but their predictive ability for SAP remains unexplored. We aim to testify whether CPD could be an indicator of AP severity in the early stage of the disease. METHODS The prospective observational study was conducted in the emergency department ward of a territory hospital in Shanghai. The enrolled AP patients should meet 2012 Atlanta guideline. RESULTS Totally, 103 AP patients and 62 healthy controls were enrolled and patients were classified into mild AP (n = 30), moderate SAP (n = 42), and SAP (n = 31). Forty-two CPD parameters were examined in first 3 days of admission. Four CPD parameters were highest in SAP on admission and were constantly different among 3 groups during first 3 days of hospital stay. Eighteen CPD parameters were found correlated with the occurrence of SAP. Stepwise multivariate logistic regression analysis identified a scoring system of 4 parameters (SD_LALS_NE, MN_LALS_LY, SD_LMALS_MO, and SD_AL2_MO) with a sensitivity of 96.8%, specificity of 65.3%, and AUC of 0.87 for diagnostic accuracy on early identification of SAP. AUC of this scoring system was comparable with MCTSI, SOFA, APACHE II, MMS, BISAP, or biomarkers as CRP, PCT, and WBC in prediction of SAP and ICU transfer or death. CONCLUSIONS Several leukocyte CPD parameters have been identified different among MAP, MSAP, and SAP. They might be ultimately incorporated into a predictive system marker for severity of AP.
Collapse
Affiliation(s)
- Yihui Wang
- Department of EmergencyRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Zhihong Xu
- Department of EmergencyRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yuhua Zhou
- Department of EmergencyRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Mengqi Xie
- Department of EmergencyRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xing Qi
- Department of EmergencyRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Zhiwei Xu
- Department of General SurgeryPancreatic Disease CenterRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Qi Cai
- Department of Laboratory MedicineRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Huiqiu Sheng
- Department of EmergencyRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Erzhen Chen
- Department of EmergencyRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Bing Zhao
- Department of EmergencyRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Enqiang Mao
- Department of EmergencyRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| |
Collapse
|
23
|
Transcriptome view of a killer: African swine fever virus. Biochem Soc Trans 2021; 48:1569-1581. [PMID: 32725217 PMCID: PMC7458399 DOI: 10.1042/bst20191108] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 07/01/2020] [Accepted: 07/03/2020] [Indexed: 02/07/2023]
Abstract
African swine fever virus (ASFV) represents a severe threat to global agriculture with the world's domestic pig population reduced by a quarter following recent outbreaks in Europe and Asia. Like other nucleocytoplasmic large DNA viruses, ASFV encodes a transcription apparatus including a eukaryote-like RNA polymerase along with a combination of virus-specific, and host-related transcription factors homologous to the TATA-binding protein (TBP) and TFIIB. Despite its high impact, the molecular basis and temporal regulation of ASFV transcription is not well understood. Our lab recently applied deep sequencing approaches to characterise the viral transcriptome and gene expression during early and late ASFV infection. We have characterised the viral promoter elements and termination signatures, by mapping the RNA-5' and RNA-3' termini at single nucleotide resolution. In this review, we discuss the emerging field of ASFV transcripts, transcription, and transcriptomics.
Collapse
|
24
|
Oh T, Do DT, Vo HV, Kwon HI, Lee SC, Kim MH, Nguyen DTT, Le QTV, Tran TM, Nguyen TT, Lee JY, Chae C. The Isolation and Replication of African Swine Fever Virus in Primary Renal-Derived Swine Macrophages. Front Vet Sci 2021; 8:645456. [PMID: 33816588 PMCID: PMC8017199 DOI: 10.3389/fvets.2021.645456] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 02/26/2021] [Indexed: 02/04/2023] Open
Abstract
African swine fever virus (ASFV) causes hemorrhagic disease in domestic pigs by replicating mainly in monocyte/macrophage lineages. Various primary cells including pulmonary alveolar macrophages have been used for the propagation of ASFV on this account. However, ethical constraints and consistency problems exist as it is necessary to harvest same phenotype of primary cells in order to continue a study. We suggested renal-derived swine macrophages as a novel primary cell candidate to address these issues. These primary cells proved to be permissive to both cell adapted ASFV and a wild-type ASFV. Compared to the commercial cell line MA-104, the renal-derived macrophages were more suitable to isolate the field virus. The consistent molecular characteristics of the renal-derived macrophages were demonstrated by immunocytochemistry with antibodies against macrophage cell surface markers including CD163, CD172a, and Iba-1. Viral protein p30 and p72 expression in ASFV infected macrophages was confirmed by immunocytochemistry by use of specific monoclonal antibodies. We observed increase of cell-free viral DNA and infectious virus titer in infected cell supernatant in successive days-post-infection. These results demonstrated that primary renal-derived swine macrophages are useful for ASFV isolation and propagation in terms of cell phenotypes, susceptibility to the virus, and virus production.
Collapse
Affiliation(s)
- Taehwan Oh
- Department of Veterinary Pathology, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Duy Tien Do
- Faculty of Animal Sciences and Veterinary Medicine, Nong Lam University, Ho Chi Minh City, Vietnam
| | - Hung Van Vo
- Department of Animal Health, Center for Veterinary Diagnostics, Regional Animal Health Office No. 6, Ho Chi Minh City, Vietnam
| | - Hyeok-Il Kwon
- ChoongAng Vaccine Laboratories, Daejeon, South Korea
| | | | - Min Ho Kim
- ChoongAng Vaccine Laboratories, Daejeon, South Korea
| | - Dung Thi Thu Nguyen
- Faculty of Animal Sciences and Veterinary Medicine, Nong Lam University, Ho Chi Minh City, Vietnam
| | - Quang Tin Vinh Le
- Department of Animal Health, Center for Veterinary Diagnostics, Regional Animal Health Office No. 6, Ho Chi Minh City, Vietnam
| | - Tan Minh Tran
- Department of Animal Health, Center for Veterinary Diagnostics, Regional Animal Health Office No. 6, Ho Chi Minh City, Vietnam
| | - Toan Tat Nguyen
- Faculty of Animal Sciences and Veterinary Medicine, Nong Lam University, Ho Chi Minh City, Vietnam
| | - Joo Young Lee
- ChoongAng Vaccine Laboratories, Daejeon, South Korea
| | - Chanhee Chae
- Department of Veterinary Pathology, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| |
Collapse
|
25
|
Portugal R, Goatley LC, Husmann R, Zuckermann FA, Dixon LK. A porcine macrophage cell line that supports high levels of replication of OURT88/3, an attenuated strain of African swine fever virus. Emerg Microbes Infect 2021; 9:1245-1253. [PMID: 32515659 PMCID: PMC7448849 DOI: 10.1080/22221751.2020.1772675] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The main target cells for African swine fever virus (ASFV) replication in pigs are of monocyte macrophage lineage and express markers typical of the intermediate to late stages of differentiation. The lack of a porcine cell line, which accurately represents these target cells, limits research on virus host interactions and the development of live-attenuated vaccine strains. We show here that the continuously growing, growth factor dependent ZMAC-4 porcine macrophage cell line is susceptible to infection with eight different field isolates of ASFV. Replication in ZMAC-4 cells occurred with similar kinetics and to similar high titres as in primary porcine bone marrow cells. In addition we showed that twelve passages of an attenuated strain of ASFV, OURT88/3, in ZMAC-4 cells did not reduce the ability of this virus to induce protection against challenge with virulent virus. Thus, the ZMAC-4 cells provide an alternative to primary cells for ASFV replication.
Collapse
Affiliation(s)
| | | | - Robert Husmann
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Federico A Zuckermann
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA.,Aptimmune Biologics, Inc., St Louis, MO, USA
| | | |
Collapse
|
26
|
Felipe VLJ, Paula A V, Silvio UI. Chikungunya virus infection induces differential inflammatory and antiviral responses in human monocytes and monocyte-derived macrophages. Acta Trop 2020; 211:105619. [PMID: 32634389 DOI: 10.1016/j.actatropica.2020.105619] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 07/03/2020] [Accepted: 07/03/2020] [Indexed: 01/09/2023]
Abstract
Chikungunya virus (CHIKV) is a zoonotic arthropod-borne virus that has caused several outbreaks in tropical and subtropical areas worldwide during the last 50 years. The virus is known to target different human cell types throughout the course of infection including epithelial and endothelial cells, fibroblasts, primary monocytes and monocyte-derived macrophages (MDMs). The two latter are phagocytic cell populations of the innate immune system which are involved in some aspects of CHIKV pathogenesis. However, monocytes and macrophages also potentially contribute to the control of viral replication through the expression of different pattern recognition receptors sensing viral pathogens and subsequently, inducing an type I interferone (IFN-I)-dependent antiviral immune response. The aim of this study was to determine the modulation of the expression of Toll-like receptors (TLRs), cytokine secretion capabilities and antiviral factor production in monocytes and MDMs following infection with CHIKV. Moreover, we sought to determine the replication kinetics of CHIKV in these two cell populations. We found that the maximum peak of CHIKV replication was observed between 18- and 24-hours post-infection (hpi), while after that the is strongly reduced. Furthermore, CHIKV infection induced the pro-inflammatory cytokine production starting from the first 6 hpi in both monocytes and MDMs, with similar kinetics but different protein levels. In contrast, the kinetics of transcriptional expression of some TLRs were different between both cell types. In addition, IFN-I, 2',5'-oligoadenylate synthetase 1 (OAS1), and double-stranded RNA-activated protein kinase R (PKR) mRNA levels were detected in response to CHIKV infection of monocytes and MDMs, resulting the highest expression levels at 48 hpi. In conclusion, our data provides evidence that CHIKV infection activates the TLR pathways in primary monocytes and MDMs, which play a crucial role in CHIKV pathogenesis and/or host defense, differentially. However, additional studies are required to determine the functional role of TLRs in monocytes and MDMs.
Collapse
Affiliation(s)
- Valdés López Juan Felipe
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia UdeA, Calle 70 No. 52-21, Medellín, Colombia
| | - Velilla Paula A
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia UdeA, Calle 70 No. 52-21, Medellín, Colombia.
| | - Urcuqui-Inchima Silvio
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia UdeA, Calle 70 No. 52-21, Medellín, Colombia.
| |
Collapse
|
27
|
[African swine fever]. Uirusu 2020; 70:15-28. [PMID: 33967108 DOI: 10.2222/jsv.70.15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
African swine fever (ASF) is a hemorrhagic infectious disease of Suids, which is endemic in sub-Saharan area of African continent. ASF is usually circulating sub-symptomatically among wild species of Suidae family, such as warthogs and bush pigs, by mediating Ornithodoros soft ticks. Domestic pigs (Sus scrofa) are, however, highly sensitive to the infection and show severe clinical signs with a high mortality rate, resulting a huge impact on pork production. Currently, there is no treatment or vaccine available. The etiological agent, ASFV, is highly resistant to environmental conditions, and resides in unheated pork meat or pork meat products for a long period, which may be a chance of its long-distance spread. Since August 2018, ASFV has been circulating in East and Southeast Asian countries and may possibly be introduced into Japan. Here, I describe the outline of the disease and the etiology of the pathogen in order to remind the importance of "awareness" and "preparedness" for the disease.
Collapse
|
28
|
The Leukocyte VCS Parameters Compared with Procalcitonin, Interleukin-6, and Soluble Hemoglobin Scavenger Receptor sCD163 for Prediction of Sepsis in Patients with Cirrhosis. DISEASE MARKERS 2019; 2019:1369798. [PMID: 31915467 PMCID: PMC6930384 DOI: 10.1155/2019/1369798] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Accepted: 10/29/2019] [Indexed: 12/26/2022]
Abstract
Background Patients with liver cirrhosis have a high risk of sepsis and a poor prognosis. Recently, a new standard for sepsis (Sepsis-3) has been proposed in the general population. The Coulter Lh 750 hematology analyzer can evaluate mean volume, conductivity, scatter, and distribution width of leukocyte. We tried to use Sepsis-3 criteria to study the diagnostic value of volume, conductivity, and scattering (VCS) parameters in sepsis and infection in patients with liver cirrhosis compared with traditional infection markers (PCT, IL-6, sCDl63). Methods A blinded, cohort study was conducted in three different ED populations within three affiliated hospitals. A total of 249 patients with liver cirrhosis were enrolled in the study. According to the “Sepsis-3” consensus criteria, clinical history, and laboratory examination, the subjects were divided into sepsis (n = 54), patients with infections (n = 95), and patients without systemic infections (n = 100). The blood samples of the patients were collected at the time of ED admission and were evaluated for the detection of sepsis. Results The differences of MNV, MNS, MMV, MMS, MLV, NDW, and MDW in the three groups were statistically significant. In the diagnosis of sepsis in patients with liver cirrhosis, the sensitivity of combined detection of MMV and MDW was 88.89%; the specificity was 74%. This sensitivity was significantly better than the 83.3% achieved using 0.97 mg/L as the cutoff for sCD163. In the diagnosis of infection in cirrhosis, the sensitivity of combination of MNV and MMS was increased to 86.32%; the specificity was 92%. The sensitivity was the same as that achieved by using 0.31 ng/mL as the cutoff value of PCT, but the specificity increased. Conclusion The leukocyte VCS parameter could be potential parameters for indicating sepsis and infection in patients with liver cirrhosis. The combined detection of MMV and MDW seemed to be helpful for the diagnosis of sepsis in these patients, and the combination of MNV and MMS could better indicate infection for them.
Collapse
|
29
|
Dixon LK, Islam M, Nash R, Reis AL. African swine fever virus evasion of host defences. Virus Res 2019; 266:25-33. [PMID: 30959069 PMCID: PMC6505686 DOI: 10.1016/j.virusres.2019.04.002] [Citation(s) in RCA: 122] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 03/29/2019] [Accepted: 04/04/2019] [Indexed: 12/24/2022]
Abstract
African swine fever virus causes a haemorrhagic fever in domestic pigs and wild boar. The continuing spread in Africa, Europe and Asia threatens the global pig industry. The lack of a vaccine limits disease control. To underpin rational strategies for vaccine development improved knowledge is needed of how the virus interacts with and modulates the host's responses to infection. The virus long double-stranded DNA genome codes for more than 160 proteins of which many are non-essential for replication in cells but can have important roles in evading the host's defences. Here we review knowledge of the pathways targeted by ASFV and the mechanisms by which these are inhibited. The impact of deleting single or multiple ASFV genes on virus replication in cells and infection in pigs is summarised providing information on strategies for rational development of modified live vaccines.
Collapse
Affiliation(s)
- L K Dixon
- The Pirbright Institute, Ash Road, Pirbright, Woking, Surrey, GU24 0NF, UK.
| | - M Islam
- The Pirbright Institute, Ash Road, Pirbright, Woking, Surrey, GU24 0NF, UK
| | - R Nash
- The Pirbright Institute, Ash Road, Pirbright, Woking, Surrey, GU24 0NF, UK
| | - A L Reis
- The Pirbright Institute, Ash Road, Pirbright, Woking, Surrey, GU24 0NF, UK
| |
Collapse
|
30
|
Dei Giudici S, Franzoni G, Bonelli P, Bacciu D, Sanna G, Angioi PP, Ledda M, Pilo G, Nicolussi P, Oggiano A. Interaction of historical and modern Sardinian African swine fever viruses with porcine and wild-boar monocytes and monocyte-derived macrophages. Arch Virol 2019; 164:739-745. [PMID: 30631959 PMCID: PMC6394708 DOI: 10.1007/s00705-018-04140-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 12/24/2018] [Indexed: 12/20/2022]
Abstract
African swine fever (ASF) is a contagious viral disease of wild and domestic pigs that is present in many parts of Africa, Asia and Europe, including Sardinia (Italy). Deletions in the EP402R and B602L genes have been found in almost all ASF virus (ASFV) strains circulating in Sardinia from 1990 onwards, and modern Sardinian strains (isolated after 1990) might have acquired some selective advantage compared to historical ones (isolated before 1990). Here, we analysed the host cell responses of wild boars and domestic pigs upon infection with virus variants. Higher intracellular levels of the late protein p72 were detected after infection with the modern strain 22653/14 compared to the historical strain Nu81.2, although both isolates grew at the same rate in both monocytes and monocyte-derived macrophages. Higher cytokine levels in the supernatants of ASFV-infected pig monocytes compared to pig macrophages and wild-boar cells were detected, with no differences between isolates.
Collapse
Affiliation(s)
- Silvia Dei Giudici
- Istituto Zooprofilattico Sperimentale della Sardegna, Via Vienna 2, 07100, Sassari, Italy.
| | - Giulia Franzoni
- Istituto Zooprofilattico Sperimentale della Sardegna, Via Vienna 2, 07100, Sassari, Italy
| | - Piero Bonelli
- Istituto Zooprofilattico Sperimentale della Sardegna, Via Vienna 2, 07100, Sassari, Italy
| | - Donatella Bacciu
- Istituto Zooprofilattico Sperimentale della Sardegna, Via Vienna 2, 07100, Sassari, Italy
| | - Giovanna Sanna
- Istituto Zooprofilattico Sperimentale della Sardegna, Via Vienna 2, 07100, Sassari, Italy
| | - Pier Paolo Angioi
- Istituto Zooprofilattico Sperimentale della Sardegna, Via Vienna 2, 07100, Sassari, Italy
| | - Mauro Ledda
- Department of Veterinary Medicine, University of Sassari, Via Vienna 2, 07100, Sassari, Italy
| | - Giovannantonio Pilo
- Istituto Zooprofilattico Sperimentale della Sardegna, Via Vienna 2, 07100, Sassari, Italy
| | - Paola Nicolussi
- Istituto Zooprofilattico Sperimentale della Sardegna, Via Vienna 2, 07100, Sassari, Italy
| | - Annalisa Oggiano
- Istituto Zooprofilattico Sperimentale della Sardegna, Via Vienna 2, 07100, Sassari, Italy
| |
Collapse
|
31
|
Infection, modulation and responses of antigen-presenting cells to African swine fever viruses. Virus Res 2018; 258:73-80. [PMID: 30316802 DOI: 10.1016/j.virusres.2018.10.007] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 09/05/2018] [Accepted: 10/10/2018] [Indexed: 01/01/2023]
Abstract
African swine fever (ASF) is a devastating viral disease of domestic pigs and wild boar for which there is no vaccine available. The aetiological agent ASF virus (ASFV) has a predilection for cells of the myeloid lineage. Macrophages provide a first line defence against pathogens and are the main target of ASFV, thus several studies analysed their response to infection in terms of cytokine/chemokine expression and modulation of functionality. These studies have typically used macrophages differentiated in vitro from blood or bone marrow progenitors and few studies have focused on responses of polarized macrophages (M1, M2) or functional macrophage subsets isolated from different tissues. ASFV can also infect dendritic cells (DC), but regardless of their central role in the induction of adaptive immune responses, their role in ASFV infection was only partially analysed. Future studies on ASFV-DC interaction are needed, which should take into consideration the heterogeneity within this family, composed of different subsets whose phenotype is also organ specific. Other porcine immune cells such as γδ-T cells, NK cells and fibrocytes, can act as 'non-conventional' antigen-presenting cells (APCs). In particular, γδ-T cells from ASFV immune pigs were shown to present viral antigens to T cells, but no studies have further explored the interaction of ASFV with this cell type or other non-conventional APCs. In this review we will provide an overview of the interaction of APCs with ASFV, describing the differences between virulent and attenuated strains, and suggesting areas for possible future studies.
Collapse
|
32
|
Valdés López JF, Urcuqui-Inchima S. Synergism between phorbol-12-myristate-13-acetate and vitamin D3 in the differentiation of U937 cells to monocytes and macrophages. Morphologie 2018; 102:205-218. [PMID: 30075941 DOI: 10.1016/j.morpho.2018.06.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 04/17/2018] [Accepted: 06/28/2018] [Indexed: 06/08/2023]
Abstract
Phorbol-12-myristate-13-acetate (PMA) and 1,25-dihydroxyvitamin D3 (VD3) are stimuli commonly used to induce macrophage differentiation in monocytic cell lines, but the extent of differentiation in comparison to primary tissue macrophages is unclear. Here, we examine the morphological/phenotypic markers associated with differentiation of U937 cells into monocytes/macrophages, in response to PMA or VD3 treatment. PMA stimulus but not with VD3, induced changes in cell morphology indicative of differentiation, but did not show differentiation comparable to monocyte-derive macrophage (MDM). The cells treated with PMA+VD3 for 2 days (d) acquired morphological/phenotypic features similar to those acquired by monocytes. In contrast, U937 cells treated for 2d with PMA and VD3 followed by 6d of resting in culture without PMA but in the presence of VD3 acquired morphological and phenotypic markers similar to those of MDM; i.e. reduced nucleus/cytoplasmic ratio, high auto-fluorescence and cytoplasmic complexity. Furthermore, low expression of CD14/TLR2 and high expression of CD68/CD86 were observed. In conclusion, our results indicate a synergistic effect between PMA and VD3 in U937 cells differentiation into both monocytes or macrophages and we propose a modified PMA differentiation protocol to enhance monocyte/macrophage differentiation of U937 cells.
Collapse
Affiliation(s)
- J F Valdés López
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia (UdeA), Calle 70 No. 52-21, Medellín, Colombia.
| | - S Urcuqui-Inchima
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia (UdeA), Calle 70 No. 52-21, Medellín, Colombia.
| |
Collapse
|
33
|
Sautter CA, Auray G, Python S, Liniger M, Summerfield A. Phenotypic and functional modulations of porcine macrophages by interferons and interleukin-4. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2018; 84:181-192. [PMID: 29408047 DOI: 10.1016/j.dci.2018.01.018] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 01/25/2018] [Accepted: 01/26/2018] [Indexed: 06/07/2023]
Abstract
Considering that macrophage functions are strongly impacted by the local tissue environment and the type of immune response, the aim of this study was to carefully set the methodological baseline for phenotype and functions of polarized porcine monocyte-derived macrophages. To this end, macrophages were generated in autologous serum alone or with colony-stimulating factor (CSF)-1 or CSF-2, and subsequently polarized with interferon (IFN)γ, interleukin-4 or IFNβ. IFNγ promoted expression of MHC class I, MHC class II, CD11a, and CD40 as well as LPS-induced IL-6 and IL-12. A hallmark of interleukin-4 was Arginase 1 and CD203a upregulation, without abrogating pro-inflammatory cytokine production. IFNβ induced CD169, MHC class I, CD40, CD80/86, but suppressed IL-6, IL-12 and tumor-necrosis-factor secretion. CSF-2 alone altered macrophage differentiation and promoted an IFNγ-like polarization. Altogether, the results provide a comprehensive overview of porcine macrophage polarization, and demonstrate commonalities with other species as well as peculiarities of the pig.
Collapse
Affiliation(s)
- Carmen A Sautter
- Institute of Virology and Immunology IVI, Sensemattstrasse 293, 3147, Mittelhäusern, Switzerland; Graduate School for Cellular and Biomedical Sciences, University of Bern, Freiestrasse 1, 3012, Bern, Switzerland; Department of Infectious Diseases and Pathobiology (DIP), Vetsuisse Faculty, University of Bern, Länggassstrasse 122, 3012, Bern, Switzerland.
| | - Gaël Auray
- Institute of Virology and Immunology IVI, Sensemattstrasse 293, 3147, Mittelhäusern, Switzerland.
| | - Sylvie Python
- Institute of Virology and Immunology IVI, Sensemattstrasse 293, 3147, Mittelhäusern, Switzerland.
| | - Matthias Liniger
- Institute of Virology and Immunology IVI, Sensemattstrasse 293, 3147, Mittelhäusern, Switzerland.
| | - Artur Summerfield
- Institute of Virology and Immunology IVI, Sensemattstrasse 293, 3147, Mittelhäusern, Switzerland; Department of Infectious Diseases and Pathobiology (DIP), Vetsuisse Faculty, University of Bern, Länggassstrasse 122, 3012, Bern, Switzerland.
| |
Collapse
|
34
|
Tellier L, Krieger J, Brimeyer A, Coogan A, Falis A, Rinker T, Schudel A, Thomas S, Jarrett C, Willett N, Botchwey E, Temenoff J. Localized SDF-1α Delivery Increases Pro-Healing Bone Marrow-Derived Cells in the Supraspinatus Muscle Following Severe Rotator Cuff Injury. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2018; 4:92-103. [PMID: 30288396 PMCID: PMC6166879 DOI: 10.1007/s40883-018-0052-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 03/31/2018] [Indexed: 10/17/2022]
Abstract
To examine how the chemotactic agent stromal cell-derived factor-1alpha (SDF-1α) modulates the unique cellular milieu within rotator cuff muscle following tendon injury, we developed an injectable, heparin-based microparticle platform to locally present SDF-1α within the supraspinatus muscle following severe rotator cuff injury. SDF-1α loaded, degradable, N-desulfated heparin-based microparticles were fabricated, injected into a rat model of severe rotator cuff injury, and were retained for up to 7 days at the site. The resultant inflammatory cell and mesenchymal stem cell populations were analyzed compared to uninjured contralateral controls and, after 7 days, the fold-change in anti-inflammatory, M2-like macrophages (CD11b+CD68+CD163+, 4.3X fold-change) and mesenchymal stem cells (CD29+CD44+CD90+, 3.0X, respectively) was significantly greater in muscles treated with SDF-1α loaded microparticles than unloaded microparticles or injury alone. Our results indicate that SDF-1α loaded microparticles may be a novel approach to shift the cellular composition within the supraspinatus muscle and create a more pro-regenerative milieu, which may provide a platform to improve muscle repair following rotator cuff injury in the future.
Collapse
Affiliation(s)
- L.E. Tellier
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA
| | - J.R. Krieger
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA
| | - A.L. Brimeyer
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA
| | - A.C. Coogan
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA
| | - A.A. Falis
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA
| | - T.E. Rinker
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA
| | - A. Schudel
- School of Materials Science and Engineering, Georgia Institute of Technology, Atlanta, GA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA
| | - S.N. Thomas
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA
- Winship Cancer Institute, Emory University, Decatur, GA
| | - C.D. Jarrett
- Wilmington Health Orthopedic Medical Center, Wilmington, NC
- Department of Orthopedics, Emory University, Decatur, GA
| | - N.J. Willett
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA
- Department of Orthopedics, Emory University, Decatur, GA
- Atlanta Veteran’s Affairs Medical Center, Decatur, GA
| | - E.A. Botchwey
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA
| | - J.S. Temenoff
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA
| |
Collapse
|
35
|
Kehoe K, Noels H, Theelen W, De Hert E, Xu S, Verrijken A, Arnould T, Fransen E, Hermans N, Lambeir AM, Venge P, Van Gaal L, De Meester I. Prolyl carboxypeptidase activity in the circulation and its correlation with body weight and adipose tissue in lean and obese subjects. PLoS One 2018; 13:e0197603. [PMID: 29772029 PMCID: PMC5957431 DOI: 10.1371/journal.pone.0197603] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 05/04/2018] [Indexed: 12/30/2022] Open
Abstract
Background Prolyl carboxypeptidase (PRCP) is involved in the regulation of body weight, likely by hydrolysing alpha-melanocyte-stimulating hormone and apelin in the hypothalamus and in the periphery. A link between PRCP protein concentrations in plasma and metabolic disorders has been reported. In this study, we investigated the distribution of circulating PRCP activity and assessed its relation with body weight and adipose tissue in obese patients and patients who significantly lost weight. Methods PRCP activity was measured using reversed-phase high-performance liquid chromatography in different isolated blood fractions and primary human cells to investigate the distribution of circulating PRCP. PRCP activity was measured in serum of individuals (n = 75) categorized based on their body mass index (BMI < 25.0; 25.0–29.9; 30.0–39.9; ≥ 40.0 kg/m2) and the diagnosis of metabolic syndrome. Differences in serum PRCP activity were determined before and six months after weight loss, either by diet (n = 45) or by bariatric surgery (n = 24). Potential correlations between serum PRCP activity and several metabolic and biochemical parameters were assessed. Additionally, plasma PRCP concentrations were quantified using a sensitive ELISA in the bariatric surgery group. Results White blood cells and plasma contributed the most to circulating PRCP activity. Serum PRCP activity in lean subjects was 0.83 ± 0.04 U/L and increased significantly with a rising BMI (p<0.001) and decreased upon weight loss (diet, p<0.05; bariatric surgery, p<0.001). The serum PRCP activity alteration reflected body weight changes and was found to be positively correlated with several metabolic parameters, including: total, abdominal and visceral adipose tissue. Plasma PRCP concentration was found to be significantly correlated to serum PRCP activity (0.865; p<0.001). Additionally, a significant decrease (p<0.001) in plasma PRCP protein concentration (mean ± SD) before (18.2 ± 3.7 ng/mL) and 6 months after bariatric surgery (15.7 ± 2.7 ng/mL) was found. Conclusion Our novel findings demonstrate that white blood cells and plasma contributed the most to circulating PRCP activity. Additionally, we have shown that there were significant correlations between serum PRCP activity and various metabolic parameters, and that plasma PRCP concentration was significantly correlated to serum PRCP activity. These novel findings on PRCP activity in serum support further investigation of its in vivo role and involvement in several metabolic diseases.
Collapse
Affiliation(s)
- Kaat Kehoe
- Laboratory of Medical Biochemistry, Department of Pharmaceutical Sciences, University of Antwerp, Antwerp, Belgium
| | - Heidi Noels
- Institute for Molecular Cardiovascular Research, RWTH Aachen University, Aachen, Germany
| | - Wendy Theelen
- Institute for Molecular Cardiovascular Research, RWTH Aachen University, Aachen, Germany
| | - Emilie De Hert
- Laboratory of Medical Biochemistry, Department of Pharmaceutical Sciences, University of Antwerp, Antwerp, Belgium
| | - Shenguan Xu
- Section of Clinical Chemistry, Department of Medical Sciences, University of Uppsala, Uppsala, Sweden
| | - An Verrijken
- Department of Endocrinology, Diabetology and Metabolism, Antwerp University Hospital, Edegem, Belgium
- Laboratory of Experimental Medicine and Paediatrics (LEMP), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Thierry Arnould
- Laboratory of Biochemistry and Cell Biology (URBC), Namur Research Institute for Life Sciences (NARILIS), University of Namur (UNamur), Namur, Belgium
| | - Erik Fransen
- StatUa Center for Statistics, University of Antwerp, Antwerp, Belgium
| | - Nina Hermans
- Natural Products & Food Research and Analysis (NatuRA), Department of Pharmaceutical Sciences, University of Antwerp, Antwerp, Belgium
| | - Anne-Marie Lambeir
- Laboratory of Medical Biochemistry, Department of Pharmaceutical Sciences, University of Antwerp, Antwerp, Belgium
| | - Per Venge
- Section of Clinical Chemistry, Department of Medical Sciences, University of Uppsala, Uppsala, Sweden
| | - Luc Van Gaal
- Department of Endocrinology, Diabetology and Metabolism, Antwerp University Hospital, Edegem, Belgium
- Laboratory of Experimental Medicine and Paediatrics (LEMP), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Ingrid De Meester
- Laboratory of Medical Biochemistry, Department of Pharmaceutical Sciences, University of Antwerp, Antwerp, Belgium
- * E-mail:
| |
Collapse
|
36
|
Portugal R, Leitão A, Martins C. Modulation of type I interferon signaling by African swine fever virus (ASFV) of different virulence L60 and NHV in macrophage host cells. Vet Microbiol 2018. [PMID: 29519508 DOI: 10.1016/j.vetmic.2018.02.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
ASFV causes an important disease of domestic swine and wild boar. Currently no vaccine is available, highlighting the necessity to understand ASFV modulation of innate immune responses in natural host cells. With this aim, macrophage cultures enriched in SWC9 and CD163 differentiation markers were infected in parallel with high virulent ASFV/L60 and low virulent ASFV/NHV, the latter lacking MGF 360 and 505/530 genes associated with type I interferon (IFN I) control. IFN I production and signaling were studied after completion of the viral cycles. None of the viruses increased IFN I production in host cells, and accordingly, didn't cause activation of the central mediator of the pathway IRF3. However, upon stimulation by poly:IC treatment during infections, L60 and NHV similarly inhibited IFN I production. This didn't seem to depend on IRF3 modulation since its activation levels were not significantly decreased in L60 infection and were even increased in NHV's, in comparison to stimulated mock infections. The infections didn't evidently activate JAK-STAT pathway mediators STAT1 and STAT2, but did increase expression of interferon stimulated genes (ISGs), to higher levels in NHV than L60 infection. Interestingly, in presence of IFN-α, L60 but not NHV was able to decrease significantly the expression of some of the ISGs tested. Overall, both L60 and NHV were able to inhibit IFN I production in macrophages, through a mechanism not dependent on IRF3 modulation. The high virulent isolate showed however a more effective control of the downstream ISGs expression pathway.
Collapse
Affiliation(s)
- Raquel Portugal
- Centre for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, Avenida da Universidade Técnica, 1300-477 Lisboa, Portugal.
| | - Alexandre Leitão
- Centre for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, Avenida da Universidade Técnica, 1300-477 Lisboa, Portugal.
| | - Carlos Martins
- Centre for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, Avenida da Universidade Técnica, 1300-477 Lisboa, Portugal.
| |
Collapse
|
37
|
Yang H, Zhang J, Zhang X, Shi J, Pan Y, Zhou R, Li G, Li Z, Cai G, Wu Z. CD163 knockout pigs are fully resistant to highly pathogenic porcine reproductive and respiratory syndrome virus. Antiviral Res 2018; 151:63-70. [PMID: 29337166 DOI: 10.1016/j.antiviral.2018.01.004] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 12/04/2017] [Accepted: 01/08/2018] [Indexed: 11/25/2022]
Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) causes severe economic losses to current swine production worldwide. Highly pathogenic PRRSV (HP-PRRSV), originated from a genotype 2 PRRSV, is more virulent than classical PRRSV and further exacerbates the economic impact. HP-PRRSV has become the predominant circulating field strain in China since 2006. CD163 is a cellular receptor for PRRSV. The depletion of CD163 whole protein or SRCR5 region (interaction site for the virus) confers resistance to infection of several PRRSV isolates in pigs or cultured host cells. In this study, we described the generation of a CD163 knockout (KO) pig in which the CD163 protein was ablated by using CRISPR/Cas9 gene targeting and somatic cell nuclear transfer (SCNT) technologies. Challenge with HP-PRRSV TP strain showed that CD163 KO pigs are completely resistant to viral infection manifested by the absence of viremia, antibody response, high fever or any other PRRS-associated clinical signs. By comparison, wild-type (WT) controls displayed typical signs of PRRSV infection and died within 2 weeks after infection. Deletion of CD163 showed no adverse effects to the macrophages on immunophenotyping and biological activity as hemoglobin-haptoglobin scavenger. The results demonstrated that CD163 knockout confers full resistance to HP-PRRSV infection to pigs without impairing the biological function associated with the gene.
Collapse
Affiliation(s)
- Huaqiang Yang
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China; Wens Foodstuff Group Co., Ltd., Yunfu, China.
| | - Jian Zhang
- Wens Foodstuff Group Co., Ltd., Yunfu, China
| | - Xianwei Zhang
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China; Wens Foodstuff Group Co., Ltd., Yunfu, China
| | - Junsong Shi
- Wens Foodstuff Group Co., Ltd., Yunfu, China
| | - Yongfei Pan
- Wens Foodstuff Group Co., Ltd., Yunfu, China
| | - Rong Zhou
- Wens Foodstuff Group Co., Ltd., Yunfu, China
| | - Guoling Li
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Zicong Li
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Gengyuan Cai
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China; Wens Foodstuff Group Co., Ltd., Yunfu, China
| | - Zhenfang Wu
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China; Wens Foodstuff Group Co., Ltd., Yunfu, China.
| |
Collapse
|
38
|
Abstract
African swine fever (ASF) is an acute and often fatal disease affecting domestic pigs and wild boar, with severe economic consequences for affected countries. ASF is endemic in sub-Saharan Africa and the island of Sardinia, Italy. Since 2007, the virus emerged in the republic of Georgia, and since then spread throughout the Caucasus region and Russia. Outbreaks have also been reported in Belarus, Ukraine, Lithuania, Latvia, Estonia, Romania, Moldova, Czech Republic, and Poland, threatening neighboring West European countries. The causative agent, the African swine fever virus (ASFV), is a large, enveloped, double-stranded DNA virus that enters the cell by macropinocytosis and a clathrin-dependent mechanism. African Swine Fever Virus is able to interfere with various cellular signaling pathways resulting in immunomodulation, thus making the development of an efficacious vaccine very challenging. Inactivated preparations of African Swine Fever Virus do not confer protection, and the role of antibodies in protection remains unclear. The use of live-attenuated vaccines, although rendering suitable levels of protection, presents difficulties due to safety and side effects in the vaccinated animals. Several African Swine Fever Virus proteins have been reported to induce neutralizing antibodies in immunized pigs, and vaccination strategies based on DNA vaccines and recombinant proteins have also been explored, however, without being very successful. The complexity of the virus particle and the ability of the virus to modulate host immune responses are most likely the reason for this failure. Furthermore, no permanent cell lines able to sustain productive virus infection by both virulent and naturally attenuated African Swine Fever Virus strains exist so far, thus impairing basic research and the commercial production of attenuated vaccine candidates.
Collapse
|
39
|
Arias M, de la Torre A, Dixon L, Gallardo C, Jori F, Laddomada A, Martins C, Parkhouse RM, Revilla Y, Rodriguez F, Sanchez-Vizcaino JM. Approaches and Perspectives for Development of African Swine Fever Virus Vaccines. Vaccines (Basel) 2017; 5:vaccines5040035. [PMID: 28991171 PMCID: PMC5748602 DOI: 10.3390/vaccines5040035] [Citation(s) in RCA: 132] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 10/01/2017] [Accepted: 10/03/2017] [Indexed: 12/25/2022] Open
Abstract
African swine fever (ASF) is a complex disease of swine, caused by a large DNA virus belonging to the family Asfarviridae. The disease shows variable clinical signs, with high case fatality rates, up to 100%, in the acute forms. ASF is currently present in Africa and Europe where it circulates in different scenarios causing a high socio-economic impact. In most affected regions, control has not been effective in part due to lack of a vaccine. The availability of an effective and safe ASFV vaccines would support and enforce control-eradication strategies. Therefore, work leading to the rational development of protective ASF vaccines is a high priority. Several factors have hindered vaccine development, including the complexity of the ASF virus particle and the large number of proteins encoded by its genome. Many of these virus proteins inhibit the host's immune system thus facilitating virus replication and persistence. We review previous work aimed at understanding ASFV-host interactions, including mechanisms of protective immunity, and approaches for vaccine development. These include live attenuated vaccines, and "subunit" vaccines, based on DNA, proteins, or virus vectors. In the shorter to medium term, live attenuated vaccines are the most promising and best positioned candidates. Gaps and future research directions are evaluated.
Collapse
Affiliation(s)
- Marisa Arias
- European Union Reference Laboratory for ASF, Centro de Investigación en Sanidad Animal (INIA-CISA), 28015 Madrid, Spain; (A.D.L.T.); (C.G.)
- Correspondence: ; Tel.: +34-916-202-300
| | - Ana de la Torre
- European Union Reference Laboratory for ASF, Centro de Investigación en Sanidad Animal (INIA-CISA), 28015 Madrid, Spain; (A.D.L.T.); (C.G.)
| | - Linda Dixon
- The Pirbright Institute (TPI), Surrey GU24 0NF, UK;
| | - Carmina Gallardo
- European Union Reference Laboratory for ASF, Centro de Investigación en Sanidad Animal (INIA-CISA), 28015 Madrid, Spain; (A.D.L.T.); (C.G.)
| | - Ferran Jori
- ASTRE, University of Montpellier, CIRAD, INRA, F-34398 Montpellier, France
| | - Alberto Laddomada
- Istituto Zooprofilattico Sperimentale della Sardegna (IZS-Sardegna), 07100 Sassari, Sardinia, Italy;
| | - Carlos Martins
- Faculdade de Medicina Veterinária (FMV-ULisboa), 1300-477 Lisbon, Portugal;
| | - R. Michael Parkhouse
- Instituto Gulbenkian de Ciência (IGC), Rua Quinta Grande 6, 2780-156 Oeiras, Portugal;
| | - Yolanda Revilla
- Centro de Biología Molecular Severo Ochoa (CBMSO-CSIC-UAM), C/ Nicolás Cabrera nº 1, Campus de Cantoblanco, 28049 Madrid, Spain;
| | - Fernando Rodriguez
- Institute for Research and Technology Food and Agriculture (IRTA), Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Campus de la Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain;
| | - Jose-Manuel Sanchez-Vizcaino
- OIE Reference Laboratory for ASF, Centro de Vigilancia Sanitaria Veterinaria (VISAVET), Universidad Complutense de Madrid, Avda. Puerta del Hierro, 28040 Madrid, Spain;
| |
Collapse
|
40
|
Phenotyping and susceptibility of established porcine cells lines to African Swine Fever Virus infection and viral production. Sci Rep 2017; 7:10369. [PMID: 28871180 PMCID: PMC5583235 DOI: 10.1038/s41598-017-09948-x] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 08/01/2017] [Indexed: 02/07/2023] Open
Abstract
African swine fever virus (ASFV) is a highly pathogenic, double-stranded DNA virus with a marked tropism for cells of the monocyte-macrophage lineage, affecting swine species and provoking severe economic losses and health threats. In the present study, four established porcine cell lines, IPAM-WT, IPAM-CD163, C∆2+ and WSL, were compared to porcine alveolar macrophage (PAM) in terms of surface marker phenotype, susceptibility to ASFV infection and virus production. The virulent ASFV Armenia/07, E70 or the naturally attenuated NHV/P68 strains were used as viral models. Cells expressed only low levels of specific receptors linked to the monocyte/macrophage lineage, with low levels of infection overall, with the exception of WSL, which showed more efficient production of strain NHV/P68 but not of strains E70 and Armenia/07.
Collapse
|
41
|
Padilla L, Dakhel S, Adan J, Masa M, Martinez JM, Roque L, Coll T, Hervas R, Calvis C, Llinas L, Buenestado S, Castellsague J, Messeguer R, Mitjans F, Hernandez JL. S100A7: from mechanism to cancer therapy. Oncogene 2017; 36:6749-6761. [PMID: 28825725 DOI: 10.1038/onc.2017.283] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 06/02/2017] [Accepted: 07/08/2017] [Indexed: 12/21/2022]
Abstract
Within the tumor, malignant and stromal cells support each other by secreting a wide variety of growth factors and cytokines, allowing tumor growth and disease progression. The identification and regulation of those key factors in this crosstalk has opened the opportunity to develop new therapeutic strategies that not only act on the tumor cells but also on the stroma. Among these factors, S100A7 protein has gained interest in the last years. With key roles in cell motility its expression correlates with increased tumor growth, angiogenesis and metastatic potential. This work aims to deepen in the role played by extracellular S100A7 in the tumor microenvironment, offering a new integrative insight of its mechanism of action on each cellular compartment (tumor, endothelial, immune and fibroblast). As a result, we demonstrate its implication in cell migration and invasion, and its important contribution to the formation of a proinflammatory and proangiogenic environment that favors tumor progression and metastasis. Furthermore, we define its possible role in the pre-metastatic niche formation. Considering the relevance of S100A7 in cancer progression, we have developed neutralizing monoclonal antibodies, reporting for the first time the proof of principle of this promising therapeutic strategy for cancer treatment.
Collapse
Affiliation(s)
- L Padilla
- Biomed Division, LEITAT Technological Center, Barcelona, Spain
| | - S Dakhel
- Biomed Division, LEITAT Technological Center, Barcelona, Spain
| | - J Adan
- Biomed Division, LEITAT Technological Center, Barcelona, Spain
| | - M Masa
- Biomed Division, LEITAT Technological Center, Barcelona, Spain
| | - J M Martinez
- Biomed Division, LEITAT Technological Center, Barcelona, Spain
| | - L Roque
- Biomed Division, LEITAT Technological Center, Barcelona, Spain
| | - T Coll
- Biomed Division, LEITAT Technological Center, Barcelona, Spain
| | - R Hervas
- Biomed Division, LEITAT Technological Center, Barcelona, Spain
| | - C Calvis
- Biomed Division, LEITAT Technological Center, Barcelona, Spain
| | - L Llinas
- Biomed Division, LEITAT Technological Center, Barcelona, Spain
| | - S Buenestado
- Biomed Division, LEITAT Technological Center, Barcelona, Spain
| | - J Castellsague
- Biomed Division, LEITAT Technological Center, Barcelona, Spain
| | - R Messeguer
- Biomed Division, LEITAT Technological Center, Barcelona, Spain
| | - F Mitjans
- Biomed Division, LEITAT Technological Center, Barcelona, Spain
| | - J L Hernandez
- Biomed Division, LEITAT Technological Center, Barcelona, Spain
| |
Collapse
|
42
|
Sharma R, Ghasparian A, Robinson JA, McCullough KC. Dendritic Cell Sensing of Hydrophobic Di- and Triacylated Lipopeptides Self-Assembled within Synthetic Virus-like Particles. THE JOURNAL OF IMMUNOLOGY 2017. [PMID: 28630093 DOI: 10.4049/jimmunol.1600521] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Dendritic cells (DCs) play critical roles in developing immune defenses. One important aspect is interaction with pathogen-associated molecular patterns (PAMPs)/danger-associated molecular patterns, including di- and triacylated lipopeptides. Isolated or synthetic lipopeptides are potent vaccine adjuvants, interacting with cell surface TLR2 heterodimers. In contrast, deep embedment within bacteria cell walls would impair lipopeptide interaction with cell surface TLR2, requiring degradation for PAMP recognition. Accordingly, DC processing in the absence of surface TLR2 ligation was defined using synthetic virus-like particles (SVLPs) carrying hydrophobic TLR2 PAMPs within di- and triacylated lipopeptide cores (P2Cys-SVLPs and P3Cys-SVLPs) compared with SVLPs lacking immunomodulatory lipopeptides. DCs rapidly and efficiently internalized SVLPs, which was dominated by slow endocytic processing via macropinocytosis, although some caveolar endocytosis was implicated. This delivered SVLPs primarily into macropinosomes often interacting with EEA-1+ early endosomes. Although endoplasmic reticulum association was occasionally noted, association with recycling/sorting structures was not observed. Involvement of LysoTracker+ structures slowly increased with time, with SVLPs present in such structures ultimately dominating. Only SVLPs carrying di- and triacylated lipopeptide cores induced DC activation and maturation independently of surface TLR2 ligation. Intracellular recognition of SVLP TLR2 ligands was confirmed by observing SVLPs' association with internal TLR2, which had similar kinetics to SVLP association with LysoTracker. This related to inflammatory cytokine induction by SVLP+ DCs, with adaptive immune response activation ex vivo/in vivo. Importantly, particular DCs, not monocytes, recognized intracellular exposure of the TLR2 PAMPs carried by di- and triacylated SVLP cores, which indicates subset-distinct recognition of functional internal TLR2 ligands. Thus, vaccines carrying hydrophobic TLR2 ligands would interact with particular DCs for efficient induction of specific immunity in the absence of additional adjuvant.
Collapse
Affiliation(s)
- Rajni Sharma
- Institute of Virology and Immunology, 3147 Mittelhäusern, Switzerland
| | - Arin Ghasparian
- Department of Chemistry, University of Zürich, 8057 Zürich, Switzerland; and.,Virometix AG, 8952 Zurich, Switzerland
| | - John A Robinson
- Department of Chemistry, University of Zürich, 8057 Zürich, Switzerland; and
| | | |
Collapse
|
43
|
Abstract
BACKGROUND Sepsis most often presents to the ED, and delayed detection is harmful. WBC count is often used to detect sepsis, but changes in WBC count size also correspond to sepsis. We sought to determine if volume increases of circulating immune cells add value to the WBC count for early sepsis detection in the ED. METHODS A blinded, prospective cohort study was conducted in two different ED populations within a large academic hospital. RESULTS Neutrophil and monocyte volume parameters were measured in conjunction with routine CBC testing on a UniCel DxH 800 analyzer at the time of ED admission and were evaluated for the detection of sepsis. There were 1,320 subjects in the ED consecutively enrolled and categorized as control subjects (n = 879) and those with systemic inflammatory response syndrome (SIRS) (n = 203), infection (n = 140), or sepsis (n = 98). Compared with other parameters, monocyte distribution width (MDW) best discriminated sepsis from all other conditions (area under the curve [AUC], 0.79; 95% CI, 0.73-0.84; sensitivity, 0.77; specificity, 0.73; MDW threshold, 20.50), sepsis from SIRS (AUC, 0.74; 95% CI, 0.67-0.84), and severe sepsis from noninfected patients in the ED (AUC, 0.88; 95% CI, 0.75-0.99; negative predictive value, 99%). The added value of MDW to WBC count was statistically significant (AUC, 0.89 for MDW + WBC vs 0.81 for WBC alone; P < .01); a decision curve analysis also showed improved performance compared with WBC count alone. CONCLUSIONS The incorporation of MDW with WBC count is shown in this prospective cohort study to improve detection of sepsis compared with WBC count alone at the time of admission in the ED. TRIAL REGISTRY ClinicalTrials.gov; No.: NCT02232750; URL: www.clinicaltrials.gov.
Collapse
|
44
|
Franzoni G, Bonelli P, Graham SP, Anfossi AG, Dei Giudici S, Pilo G, Pittau M, Nicolussi P, Oggiano A. Comparative phenotypic and functional analyses of the effects of autologous plasma and recombinant human macrophage-colony stimulating factor (M-CSF) on porcine monocyte to macrophage differentiation. Vet Immunol Immunopathol 2017; 187:80-88. [PMID: 28494933 DOI: 10.1016/j.vetimm.2017.04.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 04/03/2017] [Accepted: 04/10/2017] [Indexed: 02/02/2023]
Abstract
Porcine monocyte-derived macrophages (moMΦ) have been employed as a model cell in numerous studies of the porcine immune system. However, the lack of a standardized method for moMΦ differentiation hampers the comparison of results coming from the use of different laboratory protocols. In this study we compared the use of varying concentrations of autologous plasma (10, 20 and 30% v/v) or recombinant human macrophage-colony stimulating factor (hM-CSF; 50, 100, and 200ng/ml) to differentiate porcine monocytes into macrophages. Changes in cell morphology and surface marker expression were assessed by confocal microscopy and flow cytometry. Macrophage differentiation was evaluated by analysing TNF-α response to LPS stimulation and determining cytokine secretion patterns under both basal conditions and after classical and alternative activation. The effects of the differentiation methods on metabolic activity and susceptibility to infection with the myelotropic African swine fever virus (ASFV) were also evaluated. Monocytes cultured using the different culture conditions tested augmented in dimension and cellular complexity, but increasing porcine plasma concentrations resulted in a dose dependent enhancement in granularity and a marked pleomorphism. As expected, CD163, MHC class II DR and CD203a expression were up-regulated in both hM-CSF (M-CSF-moMΦ) and autologous plasma cultured macrophages (AP-moMΦ), although a lower percentage of CD163+ cells were found following differentiation with high percentages of porcine plasma. We observed enhanced number of viable cells using high concentration of hM-CSF compared to porcine plasma, suggesting a proliferative effect. Irrespective of differentiation conditions, monocyte differentiation into macrophages resulted in an increased susceptibility to ASFV and yielded larger amounts of LPS-induced TNF-α. AP-moMΦ showed a higher basal release of IL-1RA compared to those cultured with hM-CSF and displayed a reduced ability to respond to classical activation, suggesting that the use of high percentages of porcine plasma led to the acquisition of a M2-like phenotype. We conclude that all the protocols tested in this study can be considered as suitable to produce porcine moMΦ, although the use of hM-CSF provides high responsiveness to M1 polarization. Since a higher phenotypic and functional inter-animal variability was observed in AP-moMΦ, we propose that the use of low concentration of hM-CSF should be adopted as the method of choice to provide a better reproducibility between experiments.
Collapse
Affiliation(s)
- Giulia Franzoni
- Department of Veterinary Medicine, University of Sassari, Sassari, 07100, Italy; Istituto Zooprofilattico Sperimentale della Sardegna, Sassari, 07100, Italy.
| | - Piero Bonelli
- Istituto Zooprofilattico Sperimentale della Sardegna, Sassari, 07100, Italy.
| | - Simon Paul Graham
- The Pirbright Institute, Ash Road, Pirbright, GU24 0NF, United Kingdom; School of Veterinary Medicine, University of Surrey, Guildford, GU2 7AL, United Kingdom
| | | | - Silvia Dei Giudici
- Istituto Zooprofilattico Sperimentale della Sardegna, Sassari, 07100, Italy
| | | | - Marco Pittau
- Department of Veterinary Medicine, University of Sassari, Sassari, 07100, Italy
| | - Paola Nicolussi
- Istituto Zooprofilattico Sperimentale della Sardegna, Sassari, 07100, Italy
| | - Annalisa Oggiano
- Istituto Zooprofilattico Sperimentale della Sardegna, Sassari, 07100, Italy
| |
Collapse
|
45
|
Franzoni G, Graham SP, Giudici SD, Bonelli P, Pilo G, Anfossi AG, Pittau M, Nicolussi PS, Laddomada A, Oggiano A. Characterization of the interaction of African swine fever virus with monocytes and derived macrophage subsets. Vet Microbiol 2016; 198:88-98. [PMID: 28062012 DOI: 10.1016/j.vetmic.2016.12.010] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Revised: 12/08/2016] [Accepted: 12/09/2016] [Indexed: 12/20/2022]
Abstract
African swine fever (ASF) is a devastating disease for which there is no vaccine available. The ASF virus (ASFV) primarily infects cells of the myeloid lineage and this tropism is thought to be crucial for disease pathogenesis. A detailed in vitro characterization of the interactions of a virulent Sardinian isolate (22653/14) and a tissue culture adapted avirulent strain (BA71V) of ASFV with porcine monocytes, un-activated (moMΦ), classically (moM1) and alternatively (moM2) activated monocyte-derived macrophages was conducted in an attempt to better understand this relationship. Using a multiplicity-of-infection (MOI) of 1, both viruses were able to infect monocytes and macrophage subsets, but BA71V presented a reduced ability to infect moM1 compared to 22653/14, with higher expression of early compared to late proteins. Using an MOI of 0.01, only 22653/14 was able to replicate in all the macrophage subsets, with initially lowest in moM1 and moM2. No differences were observed in the expression of CD163 between ASFV infected and uninfected bystander cells. ASFV down-regulated CD16 expression but did not modulate MHC class II levels in monocytes and macrophage subsets. BA71V-infected but not 22653/14-infected moMΦ and moM2 presented with a reduced expression of MHC class I compared to the mock-infected controls. Higher levels of IL-18, IL1-β and IL-1α were released from moM1 after infection with BA71V compared to 22653/14 or mock-infected control. These results revealed differences between these ASFV strains, suggesting that virulent isolates have evolved mechanisms to counteract activated macrophages responses, promoting their survival, dissemination in the host and so ASF pathogenesis.
Collapse
Affiliation(s)
- Giulia Franzoni
- Department of Veterinary Medicine, University of Sassari, Sassari, 07100, Italy; Istituto Zooprofilattico Sperimentale della Sardegna, Sassari, 07100, Italy.
| | - Simon P Graham
- School of Veterinary Medicine, University of Surrey, Guildford, GU2 7AL, United Kingdom; The Pirbright Institute, Ash Road, Pirbright, GU24 0NF, United Kingdom.
| | - Silvia Dei Giudici
- Istituto Zooprofilattico Sperimentale della Sardegna, Sassari, 07100, Italy.
| | - Piero Bonelli
- Istituto Zooprofilattico Sperimentale della Sardegna, Sassari, 07100, Italy.
| | - Giovannantonio Pilo
- Istituto Zooprofilattico Sperimentale della Sardegna, Sassari, 07100, Italy.
| | - Antonio G Anfossi
- Department of Veterinary Medicine, University of Sassari, Sassari, 07100, Italy.
| | - Marco Pittau
- Department of Veterinary Medicine, University of Sassari, Sassari, 07100, Italy.
| | - Paola S Nicolussi
- Istituto Zooprofilattico Sperimentale della Sardegna, Sassari, 07100, Italy.
| | - Alberto Laddomada
- Istituto Zooprofilattico Sperimentale della Sardegna, Sassari, 07100, Italy.
| | - Annalisa Oggiano
- Istituto Zooprofilattico Sperimentale della Sardegna, Sassari, 07100, Italy.
| |
Collapse
|
46
|
Singleton H, Graham SP, Bodman-Smith KB, Frossard JP, Steinbach F. Establishing Porcine Monocyte-Derived Macrophage and Dendritic Cell Systems for Studying the Interaction with PRRSV-1. Front Microbiol 2016; 7:832. [PMID: 27313573 PMCID: PMC4889594 DOI: 10.3389/fmicb.2016.00832] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 05/17/2016] [Indexed: 11/13/2022] Open
Abstract
Monocyte-derived macrophages (MoMØ) and monocyte-derived dendritic cells (MoDC) are two model systems well established in human and rodent systems that can be used to study the interaction of pathogens with host cells. Porcine reproductive and respiratory syndrome virus (PRRSV) is known to infect myeloid cells, such as macrophages (MØ) and dendritic cells (DC). Therefore, this study aimed to establish systems for the differentiation and characterization of MoMØ and MoDC for subsequent infection with PRRSV-1. M-CSF differentiated MoMØ were stimulated with activators for classical (M1) or alternative (M2) activation. GM-CSF and IL-4 generated MoDC were activated with the well established maturation cocktail containing PAMPs and cytokines. In addition, MoMØ and MoDC were treated with dexamethasone and IL-10, which are known immuno-suppressive reagents. Cells were characterized by morphology, phenotype, and function and porcine MØ subsets highlighted some divergence from described human counterparts, while MoDC, appeared more similar to mouse and human DCs. The infection with PRRSV-1 strain Lena demonstrated different replication kinetics between MoMØ and MoDC and within subsets of each cell type. While MoMØ susceptibility was significantly increased by dexamethasone and IL-10 with an accompanying increase in CD163/CD169 expression, MoDC supported only a minimal replication of PRRSV These findings underline the high variability in the susceptibility of porcine myeloid cells toward PRRSV-1 infection.
Collapse
Affiliation(s)
- Helen Singleton
- Virology Department, Animal and Plant Health AgencySurrey, UK; Faculty of Health and Medical Sciences, University of SurreySurrey, UK
| | - Simon P Graham
- Virology Department, Animal and Plant Health AgencySurrey, UK; Faculty of Health and Medical Sciences, University of SurreySurrey, UK
| | | | | | - Falko Steinbach
- Virology Department, Animal and Plant Health AgencySurrey, UK; Faculty of Health and Medical Sciences, University of SurreySurrey, UK
| |
Collapse
|
47
|
Comparative In Vitro and In Vivo Studies of Porcine Rotavirus G9P[13] and Human Rotavirus Wa G1P[8]. J Virol 2015; 90:142-51. [PMID: 26468523 DOI: 10.1128/jvi.02401-15] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 10/01/2015] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED The changing epidemiology of group A rotavirus (RV) strains in humans and swine, including emerging G9 strains, poses new challenges to current vaccines. In this study, we comparatively assessed the pathogenesis of porcine RV (PRV) G9P[13] and evaluated the short-term cross-protection between this strain and human RV (HRV) Wa G1P[8] in gnotobiotic pigs. Complete genome sequencing demonstrated that PRV G9P[13] possessed a human-like G9 VP7 genotype but shared higher overall nucleotide identity with historic PRV strains. PRV G9P[13] induced longer rectal virus shedding and RV RNAemia in pigs than HRV Wa G1P[8] and generated complete short-term cross-protection in pigs challenged with HRV or PRV, whereas HRV Wa G1P[8] induced only partial protection against PRV challenge. Moreover, PRV G9P[13] replicated more extensively in porcine monocyte-derived dendritic cells (MoDCs) than did HRV Wa G1P[8]. Cross-protection was likely not dependent on serum virus-neutralizing (VN) antibodies, as the heterologous VN antibody titers in the sera of G9P[13]-inoculated pigs were low. Thus, our results suggest that heterologous protection by the current monovalent G1P[8] HRV vaccine against emerging G9 strains should be evaluated in clinical and experimental studies to prevent further dissemination of G9 strains. Differences in the pathogenesis of these two strains may be partially attributable to their variable abilities to replicate and persist in porcine immune cells, including dendritic cells (DCs). Additional studies are needed to evaluate the emerging G9 strains as potential vaccine candidates and to test the susceptibility of various immune cells to infection by G9 and other common HRV/PRV genotypes. IMPORTANCE The changing epidemiology of porcine and human group A rotaviruses (RVs), including emerging G9 strains, may compromise the efficacy of current vaccines. An understanding of the pathogenesis and genetic, immunological, and biological features of the new emerging RV strains will contribute to the development of new surveillance and prevention tools. Additionally, studies of cross-protection between the newly identified emerging G9 porcine RV strains and a human G1 RV vaccine strain in a susceptible host (swine) will allow evaluation of G9 strains as potential novel vaccine candidates to be included in porcine or human vaccines.
Collapse
|
48
|
McCullough KC, Milona P, Thomann-Harwood L, Démoulins T, Englezou P, Suter R, Ruggli N. Self-Amplifying Replicon RNA Vaccine Delivery to Dendritic Cells by Synthetic Nanoparticles. Vaccines (Basel) 2014; 2:735-54. [PMID: 26344889 PMCID: PMC4494254 DOI: 10.3390/vaccines2040735] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Revised: 08/29/2014] [Accepted: 09/28/2014] [Indexed: 12/12/2022] Open
Abstract
Dendritic cells (DC) play essential roles determining efficacy of vaccine delivery with respect to immune defence development and regulation. This renders DCs important targets for vaccine delivery, particularly RNA vaccines. While delivery of interfering RNA oligonucleotides to the appropriate intracellular sites for RNA-interference has proven successful, the methodologies are identical for RNA vaccines, which require delivery to RNA translation sites. Delivery of mRNA has benefitted from application of cationic entities; these offer value following endocytosis of RNA, when cationic or amphipathic properties can promote endocytic vesicle membrane perturbation to facilitate cytosolic translocation. The present review presents how such advances are being applied to the delivery of a new form of RNA vaccine, replicons (RepRNA) carrying inserted foreign genes of interest encoding vaccine antigens. Approaches have been developed for delivery to DCs, leading to the translation of the RepRNA and encoded vaccine antigens both in vitro and in vivo. Potential mechanisms favouring efficient delivery leading to translation are discussed with respect to the DC endocytic machinery, showing the importance of cytosolic translocation from acidifying endocytic structures. The review relates the DC endocytic pathways to immune response induction, and the potential advantages for these self-replicating RNA vaccines in the near future.
Collapse
Affiliation(s)
| | - Panagiota Milona
- Institute of Virology and Immunology, CH-3147 Mittelhaeusern, Switzerland.
| | | | - Thomas Démoulins
- Institute of Virology and Immunology, CH-3147 Mittelhaeusern, Switzerland.
| | - Pavlos Englezou
- Institute of Virology and Immunology, CH-3147 Mittelhaeusern, Switzerland.
| | - Rolf Suter
- Institute of Virology and Immunology, CH-3147 Mittelhaeusern, Switzerland.
| | - Nicolas Ruggli
- Institute of Virology and Immunology, CH-3147 Mittelhaeusern, Switzerland.
| |
Collapse
|
49
|
McCullough KC, Bassi I, Milona P, Suter R, Thomann-Harwood L, Englezou P, Démoulins T, Ruggli N. Self-replicating Replicon-RNA Delivery to Dendritic Cells by Chitosan-nanoparticles for Translation In Vitro and In Vivo. MOLECULAR THERAPY. NUCLEIC ACIDS 2014; 3:e173. [PMID: 25004099 PMCID: PMC4121514 DOI: 10.1038/mtna.2014.24] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2014] [Accepted: 05/20/2014] [Indexed: 02/06/2023]
Abstract
Self-amplifying replicon RNA (RepRNA) possesses high potential for increasing antigen load within dendritic cells (DCs). The major aim of the present work was to define how RepRNA delivered by biodegradable, chitosan-based nanoparticulate delivery vehicles (nanogel-alginate (NGA)) interacts with DCs, and whether this could lead to translation of the RepRNA in the DCs. Although studies employed virus replicon particles (VRPs), there are no reports on biodegradable, nanoparticulate vehicle delivery of RepRNA. VRP studies employed cytopathogenic agents, contrary to DC requirements—slow processing and antigen retention. We employed noncytopathogenic RepRNA with NGA, demonstrating for the first time the efficiency of RepRNA association with nanoparticles, NGA delivery to DCs, and RepRNA internalization by DCs. RepRNA accumulated in vesicular structures, with patterns typifying cytosolic release. This promoted RepRNA translation, in vitro and in vivo. Delivery and translation were RepRNA concentration-dependent, occurring in a kinetic manner. Including cationic lipids with chitosan during nanoparticle formation enhanced delivery and translation kinetics, but was not required for translation of immunogenic levels in vivo. This work describes for the first time the characteristics associated with chitosan-nanoparticle delivery of self-amplifying RepRNA to DCs, leading to translation of encoded foreign genes, namely influenza virus hemagglutinin and nucleoprotein.
Collapse
Affiliation(s)
| | - Isabelle Bassi
- Institute of Virology and Immunology, Mittelhäusern, Switzerland
| | - Panagiota Milona
- Institute of Virology and Immunology, Mittelhäusern, Switzerland
| | - Rolf Suter
- 1] Institute of Virology and Immunology, Mittelhäusern, Switzerland [2] Current address: Laboratory of Experimental Biophysics, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | | | - Pavlos Englezou
- Institute of Virology and Immunology, Mittelhäusern, Switzerland
| | - Thomas Démoulins
- Institute of Virology and Immunology, Mittelhäusern, Switzerland
| | - Nicolas Ruggli
- Institute of Virology and Immunology, Mittelhäusern, Switzerland
| |
Collapse
|
50
|
Démoulins T, Milona P, McCullough KC. Alginate-coated chitosan nanogels differentially modulate class-A and class-B CpG-ODN targeting of dendritic cells and intracellular delivery. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2014; 10:1739-49. [PMID: 24941461 DOI: 10.1016/j.nano.2014.06.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Revised: 05/20/2014] [Accepted: 06/05/2014] [Indexed: 12/26/2022]
Abstract
UNLABELLED CpG-oligodeoxynucleotides (CpG-ODNs) interact with dendritic cells (DCs), but evidence is less clear for CpG-ODN admixed with or incorporated into vaccine delivery vehicles. We loaded alginate-coated chitosan-nanogels (Ng) with class-A or class-B CpG-ODN, and compared with the same CpG-ODNs free or admixed with empty Ng. Experiments were performed on both porcine and human blood DC subpopulations. Encapsulation of class-A CpG-ODN (loading into Ng) strongly reduced the CpG-ODN uptake and intracellular trafficking in the cytosol; this was associated with a marked deficiency in IFN-α induction. In contrast, encapsulation of class-B CpG-ODN increased its uptake and did not influence consistently intracellular trafficking into the nucleus. The choice of CpG-ODN class as adjuvant is thus critical in terms of how it will behave with nanoparticulate vaccine delivery vehicles. The latter can have distinctive modulatory influences on the CpG-ODN, which would require definition for different CpG-ODN and delivery vehicles prior to vaccine formulation. FROM THE CLINICAL EDITOR This basic science study investigates the role of class-A and class-B CpG-oligodeoxynucleotides loaded into alginate-coated chitosan nanogels, demonstrating differential effects between the two classes as related to the use of these nanoformulations as vaccine delivery vehicles.
Collapse
Affiliation(s)
- Thomas Démoulins
- Institute of Virology and Immunology (IVI), Mittelhäusern, Switzerland.
| | - Panagiota Milona
- Institute of Virology and Immunology (IVI), Mittelhäusern, Switzerland
| | | |
Collapse
|