1
|
Coronado L, Wang M, Bohórquez JA, Muñoz-Aguilera A, Alberch M, Martínez P, Ruggli N, Ramayo-Caldas Y, Ganges L. Gene Expression Signatures of Porcine Bone Marrow-Derived Antigen-Presenting Cells Infected with Classical Swine Fever Virus. Viruses 2025; 17:160. [PMID: 40006915 PMCID: PMC11860178 DOI: 10.3390/v17020160] [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: 10/11/2024] [Revised: 01/15/2025] [Accepted: 01/16/2025] [Indexed: 02/27/2025] Open
Abstract
For a better understanding of classical swine fever (CSF) pathogenesis, a transcriptomic analysis was performed using porcine bone marrow (BM)-derived antigen-presenting cells (APCs) infected ex vivo with two different cDNA-derived classical swine fever virus (CSFV) strains, the low-virulence Pinar de Rio (vPdR-36U) or the lethal vPdR-H30K-5U. The transcriptomic profile of vPdR-36U- or vPdR-H30K-5U-infected versus noninfected cells revealed 946 and 2643 differentially expressed genes (DEGs), respectively. The upregulation of ISG15, CXCL-10, ADAM8, and CSF1 was found after infection with vPdR-36U, which could contribute to the generation of mild CSF forms. In contrast, cells infected with the lethal vPdR-H30K-5U overexpressed the immune checkpoint molecules PD-L1, CD276, and LAG3, which are involved in T-cell exhaustion and could be associated with adaptive immunity impairment. vPdR-H30K-5U also induced increased expression of PPBP, IL-8, IL-6, ECE1, and Rab27b, which are mediators of inflammatory responses that can be involved in cytokine storms. The TNF signaling pathway, which is related to the activation and proliferation of different subsets of immune cells, including CD4+ T cells, was notably upregulated in response to the low-pathogenicity virus. The Th17, Th1, and Th2 differentiation pathways were downregulated by the highly pathogenic virus only, supporting the role of T-cell-mediated immunity in protecting against CSFV.
Collapse
Affiliation(s)
- Liani Coronado
- Institute for Research and Technology in Food and Agriculture (IRTA), Programa de Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Bellaterra, 08193 Barcelona, Spain; (L.C.); (M.W.); (J.A.B.); (A.M.-A.); (M.A.); (P.M.)
- Unitat Mixta d’Investigació IRTA-UAB en Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Bellaterra, 08193 Barcelona, Spain
- Classical Swine Fever World Organization for Animal Health (WOAH) Reference Laboratory for, IRTA-CReSA, 08193 Barcelona, Spain
| | - Miaomiao Wang
- Institute for Research and Technology in Food and Agriculture (IRTA), Programa de Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Bellaterra, 08193 Barcelona, Spain; (L.C.); (M.W.); (J.A.B.); (A.M.-A.); (M.A.); (P.M.)
- Unitat Mixta d’Investigació IRTA-UAB en Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Bellaterra, 08193 Barcelona, Spain
- Classical Swine Fever World Organization for Animal Health (WOAH) Reference Laboratory for, IRTA-CReSA, 08193 Barcelona, Spain
| | - Jose Alejandro Bohórquez
- Institute for Research and Technology in Food and Agriculture (IRTA), Programa de Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Bellaterra, 08193 Barcelona, Spain; (L.C.); (M.W.); (J.A.B.); (A.M.-A.); (M.A.); (P.M.)
- Unitat Mixta d’Investigació IRTA-UAB en Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Bellaterra, 08193 Barcelona, Spain
- Classical Swine Fever World Organization for Animal Health (WOAH) Reference Laboratory for, IRTA-CReSA, 08193 Barcelona, Spain
| | - Adriana Muñoz-Aguilera
- Institute for Research and Technology in Food and Agriculture (IRTA), Programa de Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Bellaterra, 08193 Barcelona, Spain; (L.C.); (M.W.); (J.A.B.); (A.M.-A.); (M.A.); (P.M.)
- Unitat Mixta d’Investigació IRTA-UAB en Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Bellaterra, 08193 Barcelona, Spain
- Classical Swine Fever World Organization for Animal Health (WOAH) Reference Laboratory for, IRTA-CReSA, 08193 Barcelona, Spain
- Instituto Colombiano Agropecuario (ICA), Bogotá 110911, Colombia
| | - Mònica Alberch
- Institute for Research and Technology in Food and Agriculture (IRTA), Programa de Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Bellaterra, 08193 Barcelona, Spain; (L.C.); (M.W.); (J.A.B.); (A.M.-A.); (M.A.); (P.M.)
- Unitat Mixta d’Investigació IRTA-UAB en Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Bellaterra, 08193 Barcelona, Spain
- Classical Swine Fever World Organization for Animal Health (WOAH) Reference Laboratory for, IRTA-CReSA, 08193 Barcelona, Spain
| | - Patricia Martínez
- Institute for Research and Technology in Food and Agriculture (IRTA), Programa de Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Bellaterra, 08193 Barcelona, Spain; (L.C.); (M.W.); (J.A.B.); (A.M.-A.); (M.A.); (P.M.)
- Unitat Mixta d’Investigació IRTA-UAB en Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Bellaterra, 08193 Barcelona, Spain
- Classical Swine Fever World Organization for Animal Health (WOAH) Reference Laboratory for, IRTA-CReSA, 08193 Barcelona, Spain
| | - Nicolas Ruggli
- Division of Virology, Institute of Virology and Immunology (IVI), 3147 Mittelhäusern, Switzerland;
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, 3012 Bern, Switzerland
| | - Yuliaxis Ramayo-Caldas
- Animal Breeding and Genetics Program, Institute for Research and Technology in Food and Agriculture (IRTA), Torre Marimon, 08140 Caldes de Montbui, Spain;
| | - Llilianne Ganges
- Institute for Research and Technology in Food and Agriculture (IRTA), Programa de Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Bellaterra, 08193 Barcelona, Spain; (L.C.); (M.W.); (J.A.B.); (A.M.-A.); (M.A.); (P.M.)
- Unitat Mixta d’Investigació IRTA-UAB en Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Bellaterra, 08193 Barcelona, Spain
- Classical Swine Fever World Organization for Animal Health (WOAH) Reference Laboratory for, IRTA-CReSA, 08193 Barcelona, Spain
| |
Collapse
|
2
|
Zhang L, Feng X, Chen W, Wang B, He S, Fan H, Liu D. Non-infectious immune complexes downregulate the production of interferons and tumor necrosis factor-α in primary porcine alveolar macrophages in vitro. Front Vet Sci 2024; 11:1420466. [PMID: 38962699 PMCID: PMC11221350 DOI: 10.3389/fvets.2024.1420466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Accepted: 06/04/2024] [Indexed: 07/05/2024] Open
Abstract
Porcine reproductive and respiratory syndrome (PRRS) caused by the PRRS virus (PRRSV) has been harming the pig industry worldwide for nearly 40 years. Although scientific researchers have made substantial efforts to explore PRRSV pathogenesis, the immune factors influencing PRRSV infection still need to be better understood. Infectious virus-antibody immune complexes (ICs) formed by PRRSV and sub-or non-neutralizing antibodies specific for PRRSV may significantly promote the development of PRRS by enhancing PRRSV replication through antibody-dependent enhancement. However, nothing is known about whether PRRSV infection is affected by non-infectious ICs (NICs) formed by non-pathogenic/infectious antigens and corresponding specific antibodies. Here, we found that PRRSV significantly induced the transcripts and proteins of interferon-α (IFN-α), IFN-β, IFN-γ, IFN-λ1, and tumor necrosis factor-α (TNF-α) in vitro primary porcine alveolar macrophages (PAMs) in the early stage of infection. Our results showed that NICs formed by rabbit-negative IgG (RNI) and pig anti-RNI specific IgG significantly reduced the transcripts and proteins of IFN-α, IFN-β, IFN-γ, IFN-λ1, and TNF-α in vitro PAMs and significantly elevated the transcripts and proteins of interleukine-10 (IL-10) and transforming growth factor-β1 (TGF-β1) in vitro PAMs. NICs-mediated PRRSV infection showed that NICs not only significantly decreased the induction of IFN-α, IFN-β, IFN-γ, IFN-λ1, and TNF-α by PRRSV but also significantly increased the induction of IL-10 and TGF-β1 by PRRSV and considerably enhanced PRRSV replication in vitro PAMs. Our data suggested that NICs could downregulate the production of antiviral cytokines (IFN-α/β/γ/λ1 and TNF-α) during PRRSV infection in vitro and facilitated PRRSV proliferation in its host cells by inhibiting innate antiviral immune response. This study elucidated one novel immune response to PRRSV infection, which would enhance our understanding of the pathogenesis of PRRSV.
Collapse
Affiliation(s)
| | | | | | | | | | - Hongjie Fan
- College of Animal Science, Anhui Science and Technology University, Chuzhou, China
| | - Deyi Liu
- College of Animal Science, Anhui Science and Technology University, Chuzhou, China
| |
Collapse
|
3
|
Ruedas-Torres I, Sánchez-Carvajal JM, Salguero FJ, Pallarés FJ, Carrasco L, Mateu E, Gómez-Laguna J, Rodríguez-Gómez IM. The scene of lung pathology during PRRSV-1 infection. Front Vet Sci 2024; 11:1330990. [PMID: 38566751 PMCID: PMC10985324 DOI: 10.3389/fvets.2024.1330990] [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: 10/31/2023] [Accepted: 02/22/2024] [Indexed: 04/04/2024] Open
Abstract
Porcine reproductive and respiratory syndrome (PRRS) is one of the most economically important infectious diseases for the pig industry worldwide. The disease was firstly reported in 1987 and became endemic in many countries. Since then, outbreaks caused by strains of high virulence have been reported several times in Asia, America and Europe. Interstitial pneumonia, microscopically characterised by thickened alveolar septa, is the hallmark lesion of PRRS. However, suppurative bronchopneumonia and proliferative and necrotising pneumonia are also observed, particularly when a virulent strain is involved. This raises the question of whether the infection by certain strains results in an overstimulation of the proinflammatory response and whether there is some degree of correlation between the strain involved and a particular pattern of lung injury. Thus, it is of interest to know how the inflammatory response is modulated in these cases due to the interplay between virus and host factors. This review provides an overview of the macroscopic, microscopic, and molecular pathology of PRRSV-1 strains in the lung, emphasising the differences between strains of different virulence.
Collapse
Affiliation(s)
- Inés Ruedas-Torres
- United Kingdom Health Security Agency (UKHSA Porton Down), Salisbury, United Kingdom
- Department of Anatomy and Comparative Pathology and Toxicology, Pathology and Immunology Group (UCO-PIG), UIC Zoonosis y Enfermedades Emergentes ENZOEM, International Agrifood Campus of Excellence (CeiA3), Faculty of Veterinary Medicine, University of Córdoba, Córdoba, Spain
| | - José María Sánchez-Carvajal
- Department of Anatomy and Comparative Pathology and Toxicology, Pathology and Immunology Group (UCO-PIG), UIC Zoonosis y Enfermedades Emergentes ENZOEM, International Agrifood Campus of Excellence (CeiA3), Faculty of Veterinary Medicine, University of Córdoba, Córdoba, Spain
| | | | - Francisco José Pallarés
- Department of Anatomy and Comparative Pathology and Toxicology, Pathology and Immunology Group (UCO-PIG), UIC Zoonosis y Enfermedades Emergentes ENZOEM, International Agrifood Campus of Excellence (CeiA3), Faculty of Veterinary Medicine, University of Córdoba, Córdoba, Spain
| | - Librado Carrasco
- Department of Anatomy and Comparative Pathology and Toxicology, Pathology and Immunology Group (UCO-PIG), UIC Zoonosis y Enfermedades Emergentes ENZOEM, International Agrifood Campus of Excellence (CeiA3), Faculty of Veterinary Medicine, University of Córdoba, Córdoba, Spain
| | - Enric Mateu
- Department of Animal Health and Anatomy, Autonomous University of Barcelona, Barcelona, Spain
| | - Jaime Gómez-Laguna
- Department of Anatomy and Comparative Pathology and Toxicology, Pathology and Immunology Group (UCO-PIG), UIC Zoonosis y Enfermedades Emergentes ENZOEM, International Agrifood Campus of Excellence (CeiA3), Faculty of Veterinary Medicine, University of Córdoba, Córdoba, Spain
| | - Irene Magdalena Rodríguez-Gómez
- Department of Anatomy and Comparative Pathology and Toxicology, Pathology and Immunology Group (UCO-PIG), UIC Zoonosis y Enfermedades Emergentes ENZOEM, International Agrifood Campus of Excellence (CeiA3), Faculty of Veterinary Medicine, University of Córdoba, Córdoba, Spain
| |
Collapse
|
4
|
Fabros D, Charerntantanakul W. Type I and II interferons, transcription factors and major histocompatibility complexes were enhanced by knocking down the PRRSV-induced transforming growth factor beta in monocytes co-cultured with peripheral blood lymphocytes. Front Immunol 2024; 15:1308330. [PMID: 38510257 PMCID: PMC10950996 DOI: 10.3389/fimmu.2024.1308330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 02/23/2024] [Indexed: 03/22/2024] Open
Abstract
The innate and adaptive immune responses elicited by porcine reproductive and respiratory syndrome virus (PRRSV) infection are known to be poor. This study investigates the impact of PRRSV-induced transforming growth factor beta 1 (TGFβ1) on the expressions of type I and II interferons (IFNs), transcription factors, major histocompatibility complexes (MHC), anti-inflammatory and pro-inflammatory cytokines in PRRSV-infected co-cultures of monocytes and peripheral blood lymphocytes (PBL). Phosphorothioate-modified antisense oligodeoxynucleotide (AS ODN) specific to the AUG region of porcine TGFβ1 mRNA was synthesized and successfully knocked down TGFβ1 mRNA expression and protein translation. Monocytes transfected with TGFβAS1 ODN, then simultaneously co-cultured with PBL and inoculated with either classical PRRSV-2 (cPRRSV-2) or highly pathogenic PRRSV-2 (HP-PRRSV-2) showed a significant reduction in TGFβ1 mRNA expression and a significant increase in the mRNA expressions of IFNα, IFNγ, MHC-I, MHC-II, signal transducer and activator of transcription 1 (STAT1), and STAT2. Additionally, transfection of TGFβAS1 ODN in the monocyte and PBL co-culture inoculated with cPRRSV-2 significantly increased the mRNA expression of interleukin-12p40 (IL-12p40). PRRSV-2 RNA copy numbers were significantly reduced in monocytes and PBL co-culture transfected with TGFβAS1 ODN compared to the untransfected control. The yields of PRRSV-2 RNA copy numbers in PRRSV-2-inoculated monocytes and PBL co-culture were sustained and reduced by porcine TGFβ1 (rTGFβ1) and recombinant porcine IFNα (rIFNα), respectively. These findings highlight the strategy employed by PRRSV to suppress the innate immune response through the induction of TGFβ expression. The inclusion of TGFβ as a parameter for future PRRSV vaccine and vaccine adjuvant candidates is recommended.
Collapse
|
5
|
Zhang L, Feng X, Wang H, He S, Fan H, Liu D. Antibody-dependent enhancement of porcine reproductive and respiratory syndrome virus infection downregulates the levels of interferon-gamma/lambdas in porcine alveolar macrophages in vitro. Front Vet Sci 2023; 10:1150430. [PMID: 37008366 PMCID: PMC10050554 DOI: 10.3389/fvets.2023.1150430] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 02/24/2023] [Indexed: 03/17/2023] Open
Abstract
Fc gamma receptor-mediated antibody-dependent enhancement (ADE) can promote virus invasion of target cells, sometimes exacerbating the severity of the disease. ADE may be an enormous hurdle to developing efficacious vaccines for certain human and animal viruses. ADE of porcine reproductive and respiratory syndrome virus (PRRSV) infection has been demonstrated in vivo and in vitro. However, the effect of PRRSV-ADE infection on the natural antiviral immunity of the host cells is yet to be well investigated. Specifically, whether the ADE of PRRSV infection affects the levels of type II (interferon-gamma, IFN-γ) and III (interferon-lambdas, IFN-λs) interferons (IFNs) remains unclear. In this study, our results showed that PRRSV significantly induced the secretion of IFN-γ, IFN-λ1, IFN-λ3, and IFN-λ4 in porcine alveolar macrophages (PAMs) in early infection, and weakly inhibited the production of IFN-γ, IFN-λ1, IFN-λ3, and IFN-λ4 in PAMs in late infection. Simultaneously, PRRSV infection significantly increased the transcription of interferon-stimulated gene 15 (ISG15), ISG56, and 2′, 5′-oligoadenylate synthetase 2 (OAS2) in PAMs. In addition, our results showed that PRRSV infection in PAMs via the ADE pathway not only significantly decreased the synthesis of IFN-γ, IFN-λ1, IFN-λ3, and IFN-λ4 but also significantly enhanced the generation of transforming growth factor-beta1 (TGF-β1). Our results also showed that the ADE of PRRSV infection significantly reduced the mRNAs of ISG15, ISG56, and OAS2 in PAMs. In conclusion, our studies indicated that PRRSV-ADE infection suppressed innate antiviral response by downregulating the levels of type II and III IFNs, hence facilitating viral replication in PAMs in vitro. The ADE mechanism demonstrated in the present study furthered our understanding of persistent pathogenesis following PRRSV infection mediated by antibodies.
Collapse
|
6
|
Ruedas-Torres I, Sánchez-Carvajal JM, Carrasco L, Pallarés FJ, Larenas-Muñoz F, Rodríguez-Gómez IM, Gómez-Laguna J. PRRSV-1 induced lung lesion is associated with an imbalance between costimulatory and coinhibitory immune checkpoints. Front Microbiol 2023; 13:1007523. [PMID: 36713151 PMCID: PMC9878400 DOI: 10.3389/fmicb.2022.1007523] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Accepted: 12/16/2022] [Indexed: 01/15/2023] Open
Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) induces a dysregulation on the innate and adaptive immune responses. T-cell activation requires a proper interaction and precise balance between costimulatory and coinhibitory molecules, commonly known as immune checkpoints. This study aims to evaluate the expression of immune checkpoints in lung and tracheobronchial lymph node from piglets infected with two PRRSV-1 strains of different virulence during the early stage of infection. Seventy 4-week-old piglets were grouped into three experimental groups: (i) control, (ii) 3249-infected group (low virulent strain), and (iii) Lena-infected group (virulent strain) and were euthanized at 1, 3, 6, 8, and 13 days post-infection (dpi). Lung and tracheobronchial lymph node were collected to evaluate histopathological findings, PRRSV viral load and mRNA expression of costimulatory (CD28, CD226, TNFRSF9, SELL, ICOS, and CD40) and coinhibitory (CTLA4, TIGIT, PD1/PDL1, TIM3, LAG3, and IDO1) molecules through RT-qPCR. Our findings highlight a mild increase of costimulatory molecules together with an earlier and stronger up-regulation of coinhibitory molecules in both organs from PRRSV-1-infected animals, especially in the lung from virulent Lena-infected animals. The simultaneous expression of coinhibitory immune checkpoints could work in synergy to control and limit the inflammation-induced tissue damage. Further studies should be addressed to determine the role of these molecules in later stages of PRRSV infection.
Collapse
|
7
|
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
|
8
|
Li Y, Mateu E, Díaz I. Impact of Cryopreservation on Viability, Phenotype, and Functionality of Porcine PBMC. Front Immunol 2021; 12:765667. [PMID: 34912338 PMCID: PMC8666977 DOI: 10.3389/fimmu.2021.765667] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 11/08/2021] [Indexed: 11/13/2022] Open
Abstract
The use of frozen peripheral blood mononuclear cells (PBMC) is common in immunological studies. The impact of freezing PBMC has been assessed using human and mice cells, but little information is available regarding domestic animals. In the present study, the phenotype and functionality of frozen porcine PBMC were examined. In a preliminary experiment, three freezing media: fetal bovine serum plus 10% dimethyl sulfoxide, PSC cryopreservation kit, and Cryostor CS10, were compared regarding the preservation of cell viability and the response of PBMC to mitogens after thawing. After being stored one month in liquid nitrogen, cell viability was above 89% for all freezing media. The ELISPOT IFN-gamma (IFN-γ) results in response to PHA and of IgG ELISPOT in response to R848+IL-2 were similar to those obtained using fresh PBMC. In the second set of experiments, PBMC were obtained from five pigs vaccinated against Porcine reproductive and respiratory syndrome virus (PRRSV) and then frozen using Cryostor CS10. Recovered cells were phenotyped by flow cytometry using anti-CD3, CD4, CD8, and CD21 antibodies and were used to assess the PRRSV-specific responses in a proliferation experiment, an IFN-γ ELISPOT, and an IgG ELISPOT, and compared to the results obtained with fresh cells. The antigen-specific responses of frozen cells were significantly (p<0.05) impaired in the proliferation assay, particularly for CD4/CD8 double-positive T-cells and for CD21+ cells. Freezing resulted in decreased proliferation when Con A, but not PHA, was used. In ELISPOT, cryopreservation resulted in a decreased frequency of IFN-γ-secreting cells in response to PRRSV (p<0.05) but the response to PHA was not affected. No differences were observed in the IgG ELISPOT after polyclonal activation. Taken together, cryopreservation of porcine PBMC had a significant impact on the magnitude of recall antigen responses and therefore, it may affect the response of effector/memory cells but seems not to have a major impact on naïve T-cells. These results may help to the better use of frozen porcine PBMC, and to the interpretation of the results obtained from them.
Collapse
Affiliation(s)
- Yanli Li
- Departament de Sanitat i Anatomia Animals, Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
| | - Enric Mateu
- Departament de Sanitat i Anatomia Animals, Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain.,Centre de Recerca en Sanitat Animal, Institut de Recerca en Tecnologies Agroalimentàries (IRTA-CReSA), Bellaterra, Spain.,World Organisation for Animal Health (OIE) Collaborating Centre for the Research and Control of Emerging and Re-Emerging Swine Diseases in Europe (IRTA-CReSA), Bellaterra, Spain
| | - Ivan Díaz
- Centre de Recerca en Sanitat Animal, Institut de Recerca en Tecnologies Agroalimentàries (IRTA-CReSA), Bellaterra, Spain.,World Organisation for Animal Health (OIE) Collaborating Centre for the Research and Control of Emerging and Re-Emerging Swine Diseases in Europe (IRTA-CReSA), Bellaterra, Spain
| |
Collapse
|
9
|
Ruedas-Torres I, Gómez-Laguna J, Sánchez-Carvajal JM, Larenas-Muñoz F, Barranco I, Pallarés FJ, Carrasco L, Rodríguez-Gómez IM. Activation of T-bet, FOXP3, and EOMES in Target Organs From Piglets Infected With the Virulent PRRSV-1 Lena Strain. Front Immunol 2021; 12:773146. [PMID: 34956200 PMCID: PMC8697429 DOI: 10.3389/fimmu.2021.773146] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 11/08/2021] [Indexed: 01/13/2023] Open
Abstract
Transcription factors (TFs) modulate genes involved in cell-type-specific proliferative and migratory properties, metabolic features, and effector functions. Porcine reproductive and respiratory syndrome virus (PRRSV) is one of the most important pathogen agents in the porcine industry; however, TFs have been poorly studied during the course of this disease. Therefore, we aimed to evaluate the expressions of the TFs T-bet, GATA3, FOXP3, and Eomesodermin (EOMES) in target organs (the lung, tracheobronchial lymph node, and thymus) and those of different effector cytokines (IFNG, TNFA, and IL10) and the Fas ligand (FASL) during the early phase of infection with PRRSV-1 strains of different virulence. Target organs from mock-, virulent Lena-, and low virulent 3249-infected animals humanely euthanized at 1, 3, 6, 8, and 13 days post-infection (dpi) were collected to analyze the PRRSV viral load, histopathological lesions, and relative quantification through reverse transcription quantitative PCR (RT-qPCR) of the TFs and cytokines. Animals belonging to both infected groups, but mainly those infected with the virulent Lena strain, showed upregulation of the TFs T-bet, EOMES, and FOXP3, together with an increase of the cytokine IFN-γ in target organs at the end of the study (approximately 2 weeks post-infection). These results are suggestive of a stronger polarization to Th1 cells and regulatory T cells (Tregs), but also CD4+ cytotoxic T lymphocytes (CTLs), effector CD8+ T cells, and γδT cells in virulent PRRSV-1-infected animals; however, their biological functionality should be the object of further studies.
Collapse
|
10
|
Time-series transcriptomic analysis of bronchoalveolar lavage cells from virulent and low virulent PRRSV-1-infected piglets. J Virol 2021; 96:e0114021. [PMID: 34851149 PMCID: PMC8826917 DOI: 10.1128/jvi.01140-21] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) has evolved to escape the immune surveillance for a survival advantage leading to a strong modulation of host’s immune responses and favoring secondary bacterial infections. However, limited data are available on how the immunological and transcriptional responses elicited by virulent and low-virulent PRRSV-1 strains are comparable and how they are conserved during the infection. To explore the kinetic transcriptional signature associated with the modulation of host immune response at lung level, a time-series transcriptomic analysis was performed in bronchoalveolar lavage cells upon experimental in vivo infection with two PRRSV-1 strains of different virulence, virulent subtype 3 Lena strain or the low-virulent subtype 1 3249 strain. The time-series analysis revealed overlapping patterns of dysregulated genes enriched in T-cell signaling pathways among both virulent and low-virulent strains, highlighting an upregulation of co-stimulatory and co-inhibitory immune checkpoints that were disclosed as Hub genes. On the other hand, virulent Lena infection induced an early and more marked “negative regulation of immune system process” with an overexpression of co-inhibitory receptors genes related to T-cell and NK cell functions, in association with more severe lung lesion, lung viral load, and BAL cell kinetics. These results underline a complex network of molecular mechanisms governing PRRSV-1 immunopathogenesis at lung level, revealing a pivotal role of co-inhibitory and co-stimulatory immune checkpoints in the pulmonary disease, which may have an impact on T-cell activation and related pathways. These immune checkpoints, together with the regulation of cytokine-signaling pathways, modulated in a virulence-dependent fashion, orchestrate an interplay among pro- and anti-inflammatory responses. IMPORTANCE Porcine reproductive and respiratory syndrome virus (PRRSV) is one of the major threats to swine health and global production, causing substantial economic losses. We explore the mechanisms involved in the modulation of host immune response at lung level performing a time-series transcriptomic analysis upon experimental infection with two PRRSV-1 strains of different virulence. A complex network of molecular mechanisms was revealed to control the immunopathogenesis of PRRSV-1 infection, highlighting an interplay among pro- and anti-inflammatory responses as a potential mechanism to restrict inflammation-induced lung injury. Moreover, a pivotal role of co-inhibitory and co-stimulatory immune checkpoints was evidenced, which may lead to progressive dysfunction of T cells, impairing viral clearance and leading to persistent infection, favoring as well secondary bacterial infections or viral rebound. However, further studies should be conducted to evaluate the functional role of immune checkpoints in advanced stages of PRRSV infection and explore a possible T-cell exhaustion state.
Collapse
|
11
|
Amadori M, Listorti V, Razzuoli E. Reappraisal of PRRS Immune Control Strategies: The Way Forward. Pathogens 2021; 10:pathogens10091073. [PMID: 34578106 PMCID: PMC8469074 DOI: 10.3390/pathogens10091073] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 08/06/2021] [Accepted: 08/14/2021] [Indexed: 11/16/2022] Open
Abstract
The control of porcine reproductive and respiratory syndrome (PRRS) is still a major issue worldwide in the pig farming sector. Despite extensive research efforts and the practical experience gained so far, the syndrome still severely affects farmed pigs worldwide and challenges established beliefs in veterinary virology and immunology. The clinical and economic repercussions of PRRS are based on concomitant, additive features of the virus pathogenicity, host susceptibility, and the influence of environmental, microbial, and non-microbial stressors. This makes a case for integrated, multi-disciplinary research efforts, in which the three types of contributing factors are critically evaluated toward the development of successful disease control strategies. These efforts could be significantly eased by the definition of reliable markers of disease risk and virus pathogenicity. As for the host's susceptibility to PRRSV infection and disease onset, the roles of both the innate and adaptive immune responses are still ill-defined. In particular, the overt discrepancy between passive and active immunity and the uncertain role of adaptive immunity vis-à-vis established PRRSV infection should prompt the scientific community to develop novel research schemes, in which apparently divergent and contradictory findings could be reconciled and eventually brought into a satisfactory conceptual framework.
Collapse
Affiliation(s)
- Massimo Amadori
- Italian Network of Veterinary Immunology, 25125 Brescia, Italy
- Correspondence:
| | - Valeria Listorti
- Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle d’Aosta, 16129 Genoa, Italy; (V.L.); (E.R.)
| | - Elisabetta Razzuoli
- Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle d’Aosta, 16129 Genoa, Italy; (V.L.); (E.R.)
| |
Collapse
|
12
|
Hernández J, Li Y, Mateu E. Swine Dendritic Cell Response to Porcine Reproductive and Respiratory Syndrome Virus: An Update. Front Immunol 2021; 12:712109. [PMID: 34394113 PMCID: PMC8355811 DOI: 10.3389/fimmu.2021.712109] [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: 05/19/2021] [Accepted: 07/07/2021] [Indexed: 11/17/2022] Open
Abstract
Dendritic cells (DCs) are the most potent antigen-presenting cells, unique to initiate and coordinate the adaptive immune response. In pigs, conventional DCs (cDCs), plasmacytoid DCs (pDCs), and monocyte-derived DCs (moDCs) have been described in blood and tissues. Different pathogens, such as viruses, could infect these cells, and in some cases, compromise their response. The understanding of the interaction between DCs and viruses is critical to comprehend viral immunopathological responses. Porcine reproductive and respiratory syndrome virus (PRRSV) is the most important respiratory pathogen in the global pig population. Different reports support the notion that PRRSV modulates pig immune response in addition to their genetic and antigenic variability. The interaction of PRRSV with DCs is a mostly unexplored area with conflicting results and lots of uncertainties. Among the scarce certainties, cDCs and pDCs are refractory to PRRSV infection in contrast to moDCs. Additionally, response of DCs to PRRSV can be different depending on the type of DCs and maybe is related to the virulence of the viral isolate. The precise impact of this virus-DC interaction upon the development of the specific immune response is not fully elucidated. The present review briefly summarizes and discusses the previous studies on the interaction of in vitro derived bone marrow (bm)- and moDCs, and in vivo isolated cDCs, pDCs, and moDCs with PRRSV1 and 2.
Collapse
Affiliation(s)
- Jesús Hernández
- Laboratorio de Inmunología, Centro de Investigación en Alimentación y Desarrollo, Hermosillo, Mexico
| | - Yanli Li
- Departament de Sanitat i Anatomia Animals, Facultat de Veterinària, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Enric Mateu
- Departament de Sanitat i Anatomia Animals, Facultat de Veterinària, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain.,IRTA, Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Cerdanyola del Vallès, Spain
| |
Collapse
|
13
|
Li Y, Mateu E. Interaction of Type 1 Porcine Reproductive and Respiratory Syndrome Virus With In Vitro Derived Conventional Dendritic Cells. Front Immunol 2021; 12:674185. [PMID: 34177915 PMCID: PMC8221110 DOI: 10.3389/fimmu.2021.674185] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 05/12/2021] [Indexed: 12/22/2022] Open
Abstract
The present study delineates the interaction of a typical PRRSV1.1 isolate 3267 (moderate virulence) with in vitro derived pig conventional dendritic cells, cDC1, cDC2, and a CD14+ population (designated as CD14+ DCs). cDC1 and cDC2 were not susceptible to 3267 infection, but a fraction of CD14+ DCs were infected. After exposure to the virus, all three DC types remained immature as determined by no increase of maturation molecules (MHC-I, MHC-II, CD80/86, CCR7), no release of cytokines, no modification of antigen presentation abilities, and no alteration of endocytic/phagocytic capabilities. However, when infected MARC-145 cells were used as a source of viral antigens, cDC2 and CD14+ DCs showed a significant increase in the expression of maturation molecules and substantial release of cytokines, notably IL-12/IL-23p40 (by both DC types) and IL-10 (by CD14+ DCs). To address the impact of PRRSV1 3267 on TLR3- and TLR7-mediated activation, cDC1, cDC2, and CD14+ DCs were inoculated by the virus (live or UV-inactivated) for 6 h prior to or simultaneously with the addition of poly I:C (TLR3 ligand) or gardiquimod (TLR7 ligand; not used for cDC1). Compared with using TLR ligand alone, combination with the virus did not result in any alteration to the maturation markers on all DC types but changed the cytokine response to either TLR3 or TLR7 ligand. Pre-exposure of cDC2 or CD14+ DCs to the live virus resulted in an increased production of IFN-α upon poly I:C stimulation, while pre-exposure to UV-inactivated virus tended to enhance the release of IL-10 upon gardiquimod stimulation. Simultaneous addition of the live virus and the TLR ligand either had no effect (mainly in cDC2) or impaired most of the cytokine release after gardiquimod stimulation (in CD14+ DCs). When used as antigen presenting cells, cDC2 pre-inoculated by the live virus before addition of gardiquimod impaired the proliferation of CD4–CD8– T cells. In the case of CD14+ DCs, pre-exposure to the live virus or simultaneously added with TLR3 or TLR7 ligand largely decreased the proliferation of CD4–CD8+ and CD4–CD8+ T-cell subsets. For cDC1, no significant changes were observed in cytokine responses or T-cell proliferation after poly I:C stimulation. Of note, cDC1 had a short life during in vitro culturing, for which the results obtained might be biased. Overall, exposure to PRRSV1 did not induce maturation of cDC1, cDC2, or CD14+ DCs, but modified TLR3 and TLR7-associated responses (except for cDC1), which may affect the development of adaptive immunity during PRRSV1 infection. Moreover, the sensing of infected cells was different from that of the free virus.
Collapse
Affiliation(s)
- Yanli Li
- Departament de Sanitat i Anatomia Animals, Facultat de Veterinària, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Enric Mateu
- Departament de Sanitat i Anatomia Animals, Facultat de Veterinària, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain.,IRTA, Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Bellaterra, Spain
| |
Collapse
|
14
|
Ruedas-Torres I, Rodríguez-Gómez IM, Sánchez-Carvajal JM, Guil-Luna S, Larenas-Muñoz F, Pallarés FJ, Carrasco L, Gómez-Laguna J. Up-Regulation of Immune Checkpoints in the Thymus of PRRSV-1-Infected Piglets in a Virulence-Dependent Fashion. Front Immunol 2021; 12:671743. [PMID: 34046040 PMCID: PMC8144631 DOI: 10.3389/fimmu.2021.671743] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 04/15/2021] [Indexed: 12/19/2022] Open
Abstract
Virulent porcine reproductive and respiratory syndrome virus (PRRSV) strains, such as the Lena strain, have demonstrated a higher thymus tropism than low virulent strains. Virulent PRRSV strains lead to severe thymus atrophy, which could be related to marked immune dysregulation. Impairment of T-cell functions through immune checkpoints has been postulated as a strategy executed by PRRSV to subvert the immune response, however, its role in the thymus, a primary lymphoid organ, has not been studied yet. Therefore, the goal of this study was to evaluate the expression of selected immune checkpoints (PD1/PDL1, CTLA4, TIM3, LAG3, CD200R1 and IDO1) in the thymus of piglets infected with two different PRRSV-1 strains. Thymus samples from piglets infected with the low virulent 3249 strain, the virulent Lena strain and mock-infected were collected at 1, 3, 6, 8 and 13 days post-infection (dpi) to analyze PRRSV viral load, relative quantification and immunohistochemical staining of immune checkpoints. PD1/PDL1, CTLA4, TIM3, LAG3 and IDO1 immune checkpoints were significantly up-regulated in the thymus of PRRSV infected piglets, especially in those infected with the virulent Lena strain from 6 dpi onwards. This up-regulation was associated with disease progression, high viral load and cell death. Co-expression of these molecules can affect T-cell development, maturation and selection, negatively regulating the host immune response against PRRSV.
Collapse
Affiliation(s)
- Inés Ruedas-Torres
- Department of Anatomy and Comparative Pathology and Toxicology, International Agrifood Campus of Excellence (ceiA3), Faculty of Veterinary Medicine, University of Córdoba, Córdoba, Spain
| | - Irene M Rodríguez-Gómez
- Department of Anatomy and Comparative Pathology and Toxicology, International Agrifood Campus of Excellence (ceiA3), Faculty of Veterinary Medicine, University of Córdoba, Córdoba, Spain
| | - José María Sánchez-Carvajal
- Department of Anatomy and Comparative Pathology and Toxicology, International Agrifood Campus of Excellence (ceiA3), Faculty of Veterinary Medicine, University of Córdoba, Córdoba, Spain
| | - Silvia Guil-Luna
- Maimónides Institute for Biomedical Research of Córdoba, IMIBIC, Córdoba, Spain
| | - Fernanda Larenas-Muñoz
- Department of Anatomy and Comparative Pathology and Toxicology, International Agrifood Campus of Excellence (ceiA3), Faculty of Veterinary Medicine, University of Córdoba, Córdoba, Spain
| | - Francisco J Pallarés
- Department of Anatomy and Comparative Pathology and Toxicology, International Agrifood Campus of Excellence (ceiA3), Faculty of Veterinary Medicine, University of Córdoba, Córdoba, Spain
| | - Librado Carrasco
- Department of Anatomy and Comparative Pathology and Toxicology, International Agrifood Campus of Excellence (ceiA3), Faculty of Veterinary Medicine, University of Córdoba, Córdoba, Spain
| | - Jaime Gómez-Laguna
- Department of Anatomy and Comparative Pathology and Toxicology, International Agrifood Campus of Excellence (ceiA3), Faculty of Veterinary Medicine, University of Córdoba, Córdoba, Spain
| |
Collapse
|
15
|
Sánchez-Carvajal JM, Ruedas-Torres I, Carrasco L, Pallarés FJ, Mateu E, Rodríguez-Gómez IM, Gómez-Laguna J. Activation of regulated cell death in the lung of piglets infected with virulent PRRSV-1 Lena strain occurs earlier and mediated by cleaved Caspase-8. Vet Res 2021; 52:12. [PMID: 33482914 PMCID: PMC7821682 DOI: 10.1186/s13567-020-00882-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 12/16/2020] [Indexed: 02/06/2023] Open
Abstract
PRRSV-1 virulent strains cause high fever, marked respiratory disease and severe lesions in lung and lymphoid organs. Regulated cell death (RCD), such as apoptosis, necroptosis and pyroptosis, is triggered by the host to interrupt viral replication eliminating infected cells, however, although it seems to play a central role in the immunopathogenesis of PRRSV, there are significant gaps regarding their sequence and activation upon PRRSV-infection. The present study evaluated RCD events by means of caspases expression in the lung of PRRSV-1-infected pigs and their impact on pulmonary macrophage subpopulations and lung lesion. Conventional piglets were intranasally inoculated with the virulent subtype 3 Lena strain or the low virulent subtype 1 3249 strain and euthanised at 1, 3, 6, 8 and 13 dpi. Lena-infected piglets showed severe and early lung damage with a high frequency of PRRSV-N-protein+ cells, depletion of CD163+ cells and high viral load in the lung. The number of TUNEL+ cells was significantly higher than cCasp3+ cells in Lena-infected piglets during the first week post-infection. cCasp8 and to a lesser extent cCasp9 were activated by both PRRSV-1 strains after one week post-infection together with a replenishment of both CD163+ and Arg-1+ pulmonary macrophages. These results highlight the induction of other forms of RCD beyond apoptosis, such as, necroptosis and pyroptosis during the first week post-infection followed by the activation of, mainly, extrinsic apoptosis during the second week post-infection. The recovery of CD163+ macrophages at the end of the study represents an attempt to restore pulmonary macrophage subpopulations lost during the early stages of the infection but also a macrophage polarisation into M2 macrophages.
Collapse
Affiliation(s)
- Jose María Sánchez-Carvajal
- Department of Anatomy and Comparative Pathology and Toxicology, Faculty of Veterinary Medicine, University of Córdoba, 14014, Córdoba, Spain.
| | - Inés Ruedas-Torres
- Department of Anatomy and Comparative Pathology and Toxicology, Faculty of Veterinary Medicine, University of Córdoba, 14014, Córdoba, Spain.
| | - Librado Carrasco
- Department of Anatomy and Comparative Pathology and Toxicology, Faculty of Veterinary Medicine, University of Córdoba, 14014, Córdoba, Spain
| | - Francisco José Pallarés
- Department of Anatomy and Comparative Pathology and Toxicology, Faculty of Veterinary Medicine, University of Córdoba, 14014, Córdoba, Spain
| | - Enric Mateu
- Department of Animal Health and Anatomy, Faculty of Veterinary Medicine, Autonomous University of Barcelona, 08193, Bellaterra, Spain.,Institut de Recerca i Tecnologia Agroalimentàries - Centre de Recerca en Sanitat Animal (IRTA-CReSA), Campus de la Universitat Autònoma de Barcelona, 08193, Cerdanyola del Vallès, Spain
| | - Irene Magdalena Rodríguez-Gómez
- Department of Anatomy and Comparative Pathology and Toxicology, Faculty of Veterinary Medicine, University of Córdoba, 14014, Córdoba, Spain
| | - Jaime Gómez-Laguna
- Department of Anatomy and Comparative Pathology and Toxicology, Faculty of Veterinary Medicine, University of Córdoba, 14014, Córdoba, Spain
| |
Collapse
|
16
|
Go N, Belloc C, Bidot C, Touzeau S. Why, when and how should exposure be considered at the within-host scale? A modelling contribution to PRRSv infection. MATHEMATICAL MEDICINE AND BIOLOGY-A JOURNAL OF THE IMA 2020; 36:179-206. [PMID: 29790952 DOI: 10.1093/imammb/dqy005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 04/11/2018] [Indexed: 12/25/2022]
Abstract
Understanding the impact of pathogen exposure on the within-host dynamics and its outcome in terms of infectiousness is a key issue to better understand and control the infection spread. Most experimental and modelling studies tackling this issue looked at the impact of the exposure dose on the infection probability and pathogen load, very few on the within-host immune response. Our aim was to explore the impact on the within-host response not only of the exposure dose, but also of its duration and peak, for contrasted virulence levels. We used an integrative modelling approach of the within-host dynamics at the between-cell level. We focused on the porcine reproductive and respiratory syndrome virus, a major concern for the swine industry. We quantified the impact of exposure and virulence on the viral dynamics and immune response by global sensitivity analyses and descriptive statistics. We found that the area under the viral curve, an indicator of the infection severity, was fully determined by the exposure intensity. The infection duration increased with the strain virulence and, for a given strain, exhibited a positive linear correlation with the exposure intensity logarithm and the exposure duration. Taking into account the exposure intensity is hence necessary. Besides, representing the exposure due to contacts by a single punctual dose would tend to underestimate the infection duration. As the infection severity and duration both contribute to the pig infectiousness, a prolonged exposure of the adequate intensity would be recommended in an immuno-epidemiological context.
Collapse
Affiliation(s)
- Natacha Go
- BIOEPAR, INRA, Oniris, LUNAM Université, Nantes, France.,MaIAGE, INRA, Université Paris-Saclay, Jouy-en-Josas, France
| | | | - Caroline Bidot
- MaIAGE, INRA, Université Paris-Saclay, Jouy-en-Josas, France
| | - Suzanne Touzeau
- ISA, INRA, CNRS, Université Côte d'Azur, France.,BIOCORE, Inria, INRA, CNRS, UPMC Université, Université Côte d'Azur, France
| |
Collapse
|
17
|
Ferlazzo G, Ruggeri J, Boniotti MB, Guarneri F, Barbieri I, Tonni M, Bertasio C, Alborali GL, Amadori M. In vitro Cytokine Responses to Virulent PRRS Virus Strains. Front Vet Sci 2020; 7:335. [PMID: 32760741 PMCID: PMC7373743 DOI: 10.3389/fvets.2020.00335] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 05/13/2020] [Indexed: 12/16/2022] Open
Abstract
Porcine reproductive and respiratory syndrome (PRRS) affects farmed swine causing heavy direct and indirect losses. The infections sustained by PRRS viruses (PRRSV-1 and PRRSV-2) may give rise to severe clinical cases. This highlights the issue of PRRSV pathogenicity and relevant markers thereof. Since PRRSV strains can be discriminated in terms of immunotypes, we aimed to detect possible correlates of virulence in vitro based on the profile of innate immune responses induced by strains of diverse virulence. To this purpose, 10 field PRRSV isolates were investigated in assays of innate immune response to detect possible features associated with virulence. Tumor necrosis factor-α, interleukin (IL)-1beta, IL-8, IL-10, and caspase-1 were measured in cultures of PRRSV-treated peripheral blood mononuclear cells of PRRS-naive pigs, unable to support PRRSV replication. Two reference PRRSV strains (highly pathogenic and attenuated, respectively), were included in the screening. The PRRSV strains isolated from field cases were shown to vary widely in terms of inflammatory cytokine responses in vitro, which were substantially lacking with some strains including the reference, highly pathogenic one. In particular, neither the field PRRSV isolates nor the reference highly pathogenic strain gave rise to an IL-1beta response, which was consistently induced by the attenuated strain, only. This pattern of response was reversed in an inflammatory environment, in which the attenuated strain reduced the ongoing IL-1beta response. Results indicate that some pathogenic PRRSV strains can prevent a primary inflammatory response of PBMCs, associated with reduced permissiveness of mature macrophages for PRRSV replication in later phases.
Collapse
Affiliation(s)
- Gianluca Ferlazzo
- Laboratory of Animal Welfare, Clinical Chemistry and Veterinary Immunology, Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia Romagna, Brescia, Italy
| | - Jessica Ruggeri
- Laboratory of Animal Welfare, Clinical Chemistry and Veterinary Immunology, Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia Romagna, Brescia, Italy
| | - Maria Beatrice Boniotti
- Genomics Department, Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia Romagna, Brescia, Italy
| | - Flavia Guarneri
- Laboratory of Animal Welfare, Clinical Chemistry and Veterinary Immunology, Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia Romagna, Brescia, Italy
| | - Ilaria Barbieri
- Genomics Department, Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia Romagna, Brescia, Italy
| | - Matteo Tonni
- Diagnostic Laboratory, Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia Romagna, Brescia, Italy
| | - Cristina Bertasio
- Genomics Department, Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia Romagna, Brescia, Italy
| | - Giovanni Loris Alborali
- Diagnostic Laboratory, Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia Romagna, Brescia, Italy
| | - Massimo Amadori
- Laboratory of Animal Welfare, Clinical Chemistry and Veterinary Immunology, Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia Romagna, Brescia, Italy
| |
Collapse
|
18
|
Coinfections and their molecular consequences in the porcine respiratory tract. Vet Res 2020; 51:80. [PMID: 32546263 PMCID: PMC7296899 DOI: 10.1186/s13567-020-00807-8] [Citation(s) in RCA: 134] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 06/02/2020] [Indexed: 01/15/2023] Open
Abstract
Understudied, coinfections are more frequent in pig farms than single infections. In pigs, the term “Porcine Respiratory Disease Complex” (PRDC) is often used to describe coinfections involving viruses such as swine Influenza A Virus (swIAV), Porcine Reproductive and Respiratory Syndrome Virus (PRRSV), and Porcine CircoVirus type 2 (PCV2) as well as bacteria like Actinobacillus pleuropneumoniae, Mycoplasma hyopneumoniae and Bordetella bronchiseptica. The clinical outcome of the various coinfection or superinfection situations is usually assessed in the studies while in most of cases there is no clear elucidation of the fine mechanisms shaping the complex interactions occurring between microorganisms. In this comprehensive review, we aimed at identifying the studies dealing with coinfections or superinfections in the pig respiratory tract and at presenting the interactions between pathogens and, when possible, the mechanisms controlling them. Coinfections and superinfections involving viruses and bacteria were considered while research articles including protozoan and fungi were excluded. We discuss the main limitations complicating the interpretation of coinfection/superinfection studies, and the high potential perspectives in this fascinating research field, which is expecting to gain more and more interest in the next years for the obvious benefit of animal health.
Collapse
|
19
|
Sánchez-Carvajal JM, Rodríguez-Gómez IM, Ruedas-Torres I, Larenas-Muñoz F, Díaz I, Revilla C, Mateu E, Domínguez J, Martín-Valls G, Barranco I, Pallarés FJ, Carrasco L, Gómez-Laguna J. Activation of pro- and anti-inflammatory responses in lung tissue injury during the acute phase of PRRSV-1 infection with the virulent strain Lena. Vet Microbiol 2020; 246:108744. [PMID: 32605751 PMCID: PMC7265841 DOI: 10.1016/j.vetmic.2020.108744] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 05/29/2020] [Accepted: 05/31/2020] [Indexed: 12/16/2022]
Abstract
Lena virulent strain caused an increase in sera levels of IFN-γ and IL-6. Lung viral load and PRRSV-N-protein+ cells were inversely correlated with CD163+ macrophages in the lung. CD14+ cells infiltrated interstitium to possibly replenish macrophages subsets. Lena-induced microscopic lung injury was linked to an increase of iNOS+ cells. The increase of CD200R1+ and FoxP3+ cells was associated with the course of lung injury.
Porcine reproductive and respiratory syndrome virus (PRRSV) plays a key role in porcine respiratory disease complex modulating the host immune response and favouring secondary bacterial infections. Pulmonary alveolar macrophages (PAMs) are the main cells supporting PRRSV replication, with CD163 as the essential receptor for viral infection. Although interstitial pneumonia is by far the representative lung lesion, suppurative bronchopneumonia is described for PRRSV virulent strains. This research explores the role of several immune markers potentially involved in the regulation of the inflammatory response and sensitisation of lung to secondary bacterial infections by PRRSV-1 strains of different virulence. Conventional pigs were intranasally inoculated with the virulent subtype 3 Lena strain or the low virulent subtype 1 3249 strain and euthanised at 1, 3, 6 and 8 dpi. Lena-infected pigs exhibited more severe clinical signs, macroscopic lung score and viraemia associated with an increase of IL-6 and IFN-γ in sera compared to 3249-infected pigs. Extensive areas of lung consolidation corresponding with suppurative bronchopneumonia were observed in Lena-infected pigs. Lung viral load and PRRSV-N-protein+ cells were always higher in Lena-infected animals. PRRSV-N-protein+ cells were linked to a marked drop of CD163+ macrophages. The number of CD14+ and iNOS+ cells gradually increased along PRRSV-1 infection, being more evident in Lena-infected pigs. The frequency of CD200R1+ and FoxP3+ cells peaked late in both PRRSV-1 strains, with a strong correlation between CD200R1+ cells and lung injury in Lena-infected pigs. These results highlight the role of molecules involved in the earlier and higher extent of lung lesions in piglets infected with the virulent Lena strain, pointing out the activation of routes potentially involved in the restraint of the local inflammatory response.
Collapse
Affiliation(s)
- J M Sánchez-Carvajal
- Department of Anatomy and Comparative Pathology, Faculty of Veterinary Medicine, University of Córdoba, 14014, Córdoba, Spain.
| | - I M Rodríguez-Gómez
- Department of Anatomy and Comparative Pathology, Faculty of Veterinary Medicine, University of Córdoba, 14014, Córdoba, Spain
| | - I Ruedas-Torres
- Department of Anatomy and Comparative Pathology, Faculty of Veterinary Medicine, University of Córdoba, 14014, Córdoba, Spain
| | - F Larenas-Muñoz
- Department of Anatomy and Comparative Pathology, Faculty of Veterinary Medicine, University of Córdoba, 14014, Córdoba, Spain
| | - I Díaz
- Institut de Recerca i Tecnologia Agroalimentàries - Centre de Recerca en Sanitat Animal (IRTA-CReSA), Campus de la Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain
| | - C Revilla
- Department of Biotechnology, National Institute for Agricultural and Food Research and Technology (INIA), 28040, Madrid, Spain
| | - E Mateu
- Institut de Recerca i Tecnologia Agroalimentàries - Centre de Recerca en Sanitat Animal (IRTA-CReSA), Campus de la Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain; Department of Animal Health and Anatomy, Faculty of Veterinary Medicine, Autonomous University of Barcelona, 08193, Bellaterra, Spain
| | - J Domínguez
- Department of Biotechnology, National Institute for Agricultural and Food Research and Technology (INIA), 28040, Madrid, Spain
| | - G Martín-Valls
- Department of Animal Health and Anatomy, Faculty of Veterinary Medicine, Autonomous University of Barcelona, 08193, Bellaterra, Spain
| | - I Barranco
- Department of Anatomy and Comparative Pathology, Faculty of Veterinary Medicine, University of Córdoba, 14014, Córdoba, Spain
| | - F J Pallarés
- Department of Anatomy and Comparative Pathology, Faculty of Veterinary Medicine, University of Murcia, 30100, Murcia, Spain
| | - L Carrasco
- Department of Anatomy and Comparative Pathology, Faculty of Veterinary Medicine, University of Córdoba, 14014, Córdoba, Spain
| | - J Gómez-Laguna
- Department of Anatomy and Comparative Pathology, Faculty of Veterinary Medicine, University of Córdoba, 14014, Córdoba, Spain
| |
Collapse
|
20
|
Ruedas-Torres I, Rodríguez-Gómez IM, Sánchez-Carvajal JM, Pallares FJ, Barranco I, Carrasco L, Gómez-Laguna J. Activation of the extrinsic apoptotic pathway in the thymus of piglets infected with PRRSV-1 strains of different virulence. Vet Microbiol 2020; 243:108639. [PMID: 32273018 DOI: 10.1016/j.vetmic.2020.108639] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 03/04/2020] [Accepted: 03/05/2020] [Indexed: 11/17/2022]
Abstract
In the last decade, the outbreaks caused by virulent porcine reproductive and respiratory syndrome virus (PRRSV) strains from both PRRSV-1 and PRRSV-2 have considerably increased. PRRSV is able to modulate the host's immune response through the induction of apoptosis of cells in lymphoid organs like thymus, increasing the susceptibility to secondary infectious agents. The present study aimed to compare the impact of two PRRSV-1 strains, a field low virulent strain (3249 strain) and a virulent strain (Lena strain), in the thymus of infected pigs, focusing on clinical signs, histological analysis, viraemia, thymus viral load and the study of the different routes of apoptosis phenomena by immunohistochemistry. Sera and thymus samples were collected from infected animals with 3249 strain, Lena strain and mock-infected animals at 1, 3, 6, 8 and 13 days post-infection (dpi). Lena-infected animals showed severe clinical disease, high sera and thymus viral loads with evident thymic atrophy since 6 dpi, matching with PRRSV-N protein, TUNEL and cCasp3 expression in the thymic cortex. In both infected groups, there was an increase in the number of cells expressing molecules related to the extrinsic pathway of apoptosis (cCasp8 and Fas) in cortex and medulla, showing an important role in the apoptosis induction produced in thymus of PRRSV-infected piglets. The extensive apoptosis in the thymus through this pathway would lead to a decrease in the number of mature T lymphocytes and the sustained release of viral particles, which may explain the greater severity of the clinical signs observed in Lena-infected pigs.
Collapse
Affiliation(s)
- Inés Ruedas-Torres
- Department of Anatomy and Comparative Pathology, Faculty of Veterinary Medicine, University of Córdoba, 14014, Córdoba, Spain.
| | - Irene Magdalena Rodríguez-Gómez
- Department of Anatomy and Comparative Pathology, Faculty of Veterinary Medicine, University of Córdoba, 14014, Córdoba, Spain
| | - Jose María Sánchez-Carvajal
- Department of Anatomy and Comparative Pathology, Faculty of Veterinary Medicine, University of Córdoba, 14014, Córdoba, Spain
| | - Francisco José Pallares
- Department of Anatomy and Comparative Pathology, Faculty of Veterinary Medicine, University of Murcia, 30100, Murcia, Spain
| | - Inmaculada Barranco
- Department of Anatomy and Comparative Pathology, Faculty of Veterinary Medicine, University of Córdoba, 14014, Córdoba, Spain
| | - Librado Carrasco
- Department of Anatomy and Comparative Pathology, Faculty of Veterinary Medicine, University of Córdoba, 14014, Córdoba, Spain
| | - Jaime Gómez-Laguna
- Department of Anatomy and Comparative Pathology, Faculty of Veterinary Medicine, University of Córdoba, 14014, Córdoba, Spain
| |
Collapse
|
21
|
Antibody-Mediated Porcine Reproductive and Respiratory Syndrome Virus Infection Downregulates the Production of Interferon-α and Tumor Necrosis Factor-α in Porcine Alveolar Macrophages via Fc Gamma Receptor I and III. Viruses 2020; 12:v12020187. [PMID: 32046249 PMCID: PMC7077232 DOI: 10.3390/v12020187] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 02/01/2020] [Accepted: 02/06/2020] [Indexed: 12/24/2022] Open
Abstract
Antibody-dependent enhancement (ADE) contributes to the pathogenesis of porcine reproductive and respiratory syndrome virus (PRRSV)-persistent infection. However, the mechanisms of PRRSV-ADE infection are still confusing. A clear understanding of the event upon virus infection by the ADE pathway has become crucial for developing efficient intervention of the PRRSV infection. In this study, an ADE assay showed that PRRSV-ADE infection in porcine alveolar macrophages (AMs) significantly decreased the production of interferon-α (IFN-α) and tumor necrosis factor-α (TNF-α), and significantly increased the production of interleukine-10 (IL-10). A gene knockdown assay based on small interfering RNA (siRNA) showed that both Fc gamma receptor I (FcγRI) and FcγRIII in porcine AMs were involved in PRRSV-ADE infection. An activation assay showed that specific activation of FcγRI or FcγRIII in porcine AMs during PRRSV infection not only significantly decreased the production of IFN-α and TNF-α, but also significantly increased the production of IL-10 and significantly facilitated PRRSV replication. In conclusion, our studies suggested that ADE downregulated the production of IFN-α and TNF-α in porcine AMs maybe via FcγRI and FcγRIII, thereby leading to enhanced PRRSV infection.
Collapse
|
22
|
Parra-Sánchez H, Bustamante-Córdova L, Reséndiz M, Mata-Haro V, Pinelli-Saavedra A, Hernández J. Analysis of Swine Conventional Dendritic Cells, DEC205 +CD172a +/-CADM1 +, from Blood and Spleen in Response to PRRSV and PEDV. Viruses 2019; 11:v11111001. [PMID: 31683569 PMCID: PMC6893809 DOI: 10.3390/v11111001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 10/28/2019] [Accepted: 10/30/2019] [Indexed: 12/17/2022] Open
Abstract
Conventional dendritic cells (cDCs) cannot be infected by porcine reproductive and respiratory syndrome virus (PRRSV) but respond to infection via cytokine production, indicating a possible role in initiation/regulation of the immune response against PRRSV. In this work, we evaluated the responses of splenic and blood cDCs, with DEC205+CADM1+CD172a+/− phenotype, as well as those of CD163+ cells against PRRSV and porcine epidemic diarrhea virus (PEDV). Both populations were incubated in the presence of PRRSV or PEDV with and without naïve CD3+ T cells, and cytokine responses were evaluated by qPCR and ELISA. Our results showed that cDCs, but not CD163+ cells, produced IL-12 in response to PRRSV. PEDV did not induce IL-12 production. Cocultures of cDCs and autologous naïve CD3+ cells resulted in decreased IL-12 production and low expression of IFN-γ transcripts in response to PRRSV. Interestingly, cDCs increased the proliferation of naïve T cells in the presence of PRRSV compared with that achieved with monocytes and peripheral blood mononuclear cells (PBMCs). Cocultures of CD163+ cells induced IL-10 and IL-4 expression in the presence of PRRSV and PEDV, respectively. In conclusion, cDCs can selectively produce IL-12 in response to PRRSV but poorly participate in the activation of naïve T cells.
Collapse
Affiliation(s)
- Héctor Parra-Sánchez
- Laboratorio de Inmunología, Centro de Investigación en Alimentación y Desarrollo, A.C., Kilómetro 0.6 carretera la Victoria, Hermosillo 83304, Sonora, Mexico.
| | - Lorena Bustamante-Córdova
- Laboratorio de Inmunología, Centro de Investigación en Alimentación y Desarrollo, A.C., Kilómetro 0.6 carretera la Victoria, Hermosillo 83304, Sonora, Mexico.
| | - Mónica Reséndiz
- Laboratorio de Inmunología, Centro de Investigación en Alimentación y Desarrollo, A.C., Kilómetro 0.6 carretera la Victoria, Hermosillo 83304, Sonora, Mexico.
| | - Verónica Mata-Haro
- Laboratorio de Inmunología, Centro de Investigación en Alimentación y Desarrollo, A.C., Kilómetro 0.6 carretera la Victoria, Hermosillo 83304, Sonora, Mexico.
| | - Araceli Pinelli-Saavedra
- Laboratorio de Inmunología, Centro de Investigación en Alimentación y Desarrollo, A.C., Kilómetro 0.6 carretera la Victoria, Hermosillo 83304, Sonora, Mexico.
| | - Jesús Hernández
- Laboratorio de Inmunología, Centro de Investigación en Alimentación y Desarrollo, A.C., Kilómetro 0.6 carretera la Victoria, Hermosillo 83304, Sonora, Mexico.
| |
Collapse
|
23
|
Proinflammatory Cytokine Changes in Bronchoalveolar Lavage Fluid Cells Isolated from Pigs Infected Solely with Porcine Reproductive and Respiratory Syndrome Virus or Co-infected with Swine Influenza Virus. J Vet Res 2019; 63:489-495. [PMID: 31934657 PMCID: PMC6950433 DOI: 10.2478/jvetres-2019-0063] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Accepted: 09/30/2019] [Indexed: 01/15/2023] Open
Abstract
Introduction The study evaluated the patterns of local innate immune response in bronchoalveolar lavage fluid (BALF) cells of pigs infected with porcine reproductive and respiratory syndrome virus (PRRSV) alone or co-infected with swine influenza virus (SIV). Material and Methods The study was performed on 26 seven-week-old pigs in three groups: PRRSV-infected (n = 11), PRRSV and SIV-infected (n = 11), and control (n = 4). BALF was collected post euthanasia at 2 and 4 dpi (three piglets per inoculated group) and at 21 dpi (all remaining pigs). Expression of IFN-α, IFN-γ, IL-1β, IL-6, IL-8, and IL-10 mRNA was quantified in BALF cells. PRRSV RNA was quantified in BALF samples using a commercial real-time RT-PCR kit. Results The three cytokines IFN-α, IFN-γ, and IL-1β presented significant expression changes in all experimental pigs. In PRRSV-infected animals IL-8 also did, but in co-infected subjects IL-6 and IL-10 were the additional upregulated cytokines. The highest number of differentially expressed genes was observed at 4 dpi, and significant differences in cytokine gene expression did not occur between the experimental groups at any other time point. The mean PRRSV load in the BALF of PRRSV-infected pigs was higher than that of co-infected pigs at each time point, having statistical significance only at 4 dpi. Conclusion The results of the study indicate that infection with PRRSV alone as well as with SIV interferes with innate and adaptive immune response in the infected host. They also showed that co-infection demonstrates additive effects on IL-6 and IL-10 mRNA expression levels.
Collapse
|
24
|
Rodríguez-Gómez IM, Sánchez-Carvajal JM, Pallarés FJ, Mateu E, Carrasco L, Gómez-Laguna J. Virulent Lena strain induced an earlier and stronger downregulation of CD163 in bronchoalveolar lavage cells. Vet Microbiol 2019; 235:101-109. [PMID: 31282367 DOI: 10.1016/j.vetmic.2019.06.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 06/06/2019] [Accepted: 06/12/2019] [Indexed: 12/22/2022]
Abstract
Highly virulent porcine reproductive and respiratory syndrome virus (PRRSV) strains have increasingly overwhelmed Asia and Europe in recent years. This study aims to compare the clinical signs, gross and microscopic findings as well as the expression of CD163 within live pulmonary alveolar macrophages (PAMs) from bronchoalveolar lavage fluid (BALF) of pigs experimentally infected with two PRRSV strains of different virulence. Pigs were infected with either a subtype 1 PRRSV-1 3249 strain or a subtype 3 PRRSV-1 Lena strain and consecutively euthanized at 1, 3, 6, 8 and 13 days post-inoculation. Clinical signs were reported daily and BALF and lung tissue samples were collected at the different time-points and accordingly processed for their analysis. Pigs infected with Lena strain exhibited greater clinical signs as well as gross and microscopic lung scores compared to 3249-infected pigs. A decreased frequency of PAMs from BALF was observed early in pigs infected with Lena strain. Moreover, the frequency and median fluorescence intensity (MFI) of CD163 within PAMs were much lower in Lena-infected pigs than in 3249-infected pigs. This downregulation in CD163 was also observed in lung sections after the assessment of macrophages expressing CD163 by means of immunohistochemistry. This outcome may result from the effect of PRRSV replication, PRRSV-induced inflammation, the influx of immature macrophages to restore lung homeostasis and/or the evidence of CD163low cells after CD163+ cells decrease in BALF.
Collapse
Affiliation(s)
- Irene M Rodríguez-Gómez
- Department of Anatomy and Comparative Pathology, Faculty of Veterinary Medicine, University of Córdoba, International Excellence Agrifood Campus 'ceiA3', Córdoba, Spain.
| | - José M Sánchez-Carvajal
- Department of Anatomy and Comparative Pathology, Faculty of Veterinary Medicine, University of Córdoba, International Excellence Agrifood Campus 'ceiA3', Córdoba, Spain
| | - Francisco J Pallarés
- Department of Anatomy and Comparative Pathology, Faculty of Veterinary Medicine, University of Murcia, Mare Nostrum Excellence Campus, Murcia, Spain
| | - Enric Mateu
- Department of Animal Health and Anatomy, Faculty of Veterinary Medicine, Autonomous University of Barcelona, Bellaterra, Barcelona, Spain
| | - Librado Carrasco
- Department of Anatomy and Comparative Pathology, Faculty of Veterinary Medicine, University of Córdoba, International Excellence Agrifood Campus 'ceiA3', Córdoba, Spain
| | - Jaime Gómez-Laguna
- Department of Anatomy and Comparative Pathology, Faculty of Veterinary Medicine, University of Córdoba, International Excellence Agrifood Campus 'ceiA3', Córdoba, Spain
| |
Collapse
|
25
|
Sánchez-Matamoros A, Camprodon A, Maldonado J, Pedrazuela R, Miranda J. Safety and long-lasting immunity of the combined administration of a modified-live virus vaccine against porcine reproductive and respiratory syndrome virus 1 and an inactivated vaccine against porcine parvovirus and Erysipelothrix rhusiopathiae in breeding pigs. Porcine Health Manag 2019; 5:11. [PMID: 31057805 PMCID: PMC6485153 DOI: 10.1186/s40813-019-0118-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 04/08/2019] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND In the field, vaccination schedules based on modified-live virus (MLV) vaccines administered twice in gilts and every three to four months in sows are commonly used to immunize breeding herds against porcine reproductive and respiratory virus (PRRSV). Breeding sows are repeatedly vaccinated against several other agents. Thus, the combined administration of vaccines for their simultaneous use can simplify such complex immunization schedules. Here, we evaluated the safety and long-term immunity of the authorized combined administration of a PRRSV MLV vaccine and an inactivated vaccine against porcine parvovirus (PPV) and Erysipelothrix rhusiopathiae for their simultaneous use.Six-month-old naïve healthy gilts were vaccinated at day 0 and revaccinated at days 21 and 147, mimicking the abovementioned vaccination schedule. Systemic and local reactions, as well as body temperature, were measured. The excretion of PRRSV1 MLV was evaluated in oral fluids. Humoral responses against the three antigens were measured by ELISA. For PRRSV, homologous neutralizing antibodies (NAs) and homologous and heterologous cell-mediated immunity (CMI) were also assessed. RESULTS The combined administration of the tested vaccines, applied according to the manufacturer's instructions, was safe based on all evaluated parameters. Overall, we detected antibodies against PPV and PRRSV in all vaccinated pigs already after the first vaccination, whereas antibodies against E. rhusiopathiae were observed in all animals after revaccination. After subsequent revaccinations, we observed boosts for the humoral response for PPV at days 28 and 154 and at day 154 for E. rhusiopathiae. No boosts were detected during the experiment by PRRSV ELISA. In all vaccinated animals, homologous NAs against MLV were already detected before revaccination (day 21). After revaccination, there was a boost with mean titres of homologous NAs remaining constant thereafter. Concerning CMI, PRRSV-specific IFN-γ-secreting cells were already detected at day 21 for all evaluated strains and we observed boosts for all PRRSV1 strains after revaccination and recall revaccination. CONCLUSIONS We showed that the combined administration of tested vaccines described here using a vaccination schedule against PRRSV commonly implemented for breeding pigs in the field is safe and induces long-lasting humoral and cellular immunity against PRRSV, PPV, and E. rhusiopathiae.
Collapse
|
26
|
Nedumpun T, Techakriengkrai N, Thanawongnuwech R, Suradhat S. Negative Immunomodulatory Effects of Type 2 Porcine Reproductive and Respiratory Syndrome Virus-Induced Interleukin-1 Receptor Antagonist on Porcine Innate and Adaptive Immune Functions. Front Immunol 2019; 10:579. [PMID: 30972072 PMCID: PMC6443931 DOI: 10.3389/fimmu.2019.00579] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 03/04/2019] [Indexed: 12/22/2022] Open
Abstract
Impaired innate and adaptive immune responses are evidenced throughout the course of PRRSV infection. We previously reported that interleukin-1 receptor antagonist (IL-1Ra) was involved in PRRSV-induced immunosuppression during an early phase of infection. However, the exact mechanism associated with PRRSV-induced IL-1Ra immunomodulation remains unknown. To explore the immunomodulatory properties of PRRSV-induced IL-1Ra on porcine immune functions, monocyte-derived dendritic cells (MoDC) and leukocytes were cultured with type 2 PRRSV, and the immunological role of IL-1Ra was assessed by addition of anti-porcine IL-1Ra Ab. The results demonstrated that PRRSV-induced IL-1Ra reduced phagocytosis, surface expression of MHC II (SLA-DR) and CD86, as well as downregulation of IFNA and IL1 gene expression in the MoDC culture system. Interestingly, IL-1Ra secreted by the PRRSV-infected MoDC also inhibited T lymphocyte differentiation and proliferation, but not IFN-γ production. Although PRRSV-induced IL-1Ra was not directly linked to IL-10 production, it contributed to the differentiation of regulatory T lymphocytes (Treg) within the culture system. Taken together, our results demonstrated that PRRSV-induced IL-1Ra downregulates innate immune functions, T lymphocyte differentiation and proliferation, and influences collectively with IL-10 in the Treg induction. The immunomodulatory roles of IL-1Ra elucidated in this study increase our understanding of the immunobiology of PRRSV.
Collapse
Affiliation(s)
- Teerawut Nedumpun
- Interdisciplinary Program in Medical Microbiology, Graduate School, Chulalongkorn University, Bangkok, Thailand
| | - Navapon Techakriengkrai
- Department of Veterinary Microbiology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand.,Center of Excellence in Emerging Infectious Diseases in Animals, Chulalongkorn University (CU-EIDAs), Bangkok, Thailand
| | - Roongroje Thanawongnuwech
- Center of Excellence in Emerging Infectious Diseases in Animals, Chulalongkorn University (CU-EIDAs), Bangkok, Thailand.,Department of Veterinary Pathology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
| | - Sanipa Suradhat
- Department of Veterinary Microbiology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand.,Center of Excellence in Emerging Infectious Diseases in Animals, Chulalongkorn University (CU-EIDAs), Bangkok, Thailand
| |
Collapse
|
27
|
Cellular Innate Immunity against PRRSV and Swine Influenza Viruses. Vet Sci 2019; 6:vetsci6010026. [PMID: 30862035 PMCID: PMC6466325 DOI: 10.3390/vetsci6010026] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 02/21/2019] [Accepted: 02/27/2019] [Indexed: 12/11/2022] Open
Abstract
Porcine respiratory disease complex (PRDC) is a polymicrobial syndrome that results from a combination of infectious agents, such as environmental stressors, population size, management strategies, age, and genetics. PRDC results in reduced performance as well as increased mortality rates and production costs in the pig industry worldwide. This review focuses on the interactions of two enveloped RNA viruses—porcine reproductive and respiratory syndrome virus (PRRSV) and swine influenza virus (SwIV)—as major etiological agents that contribute to PRDC within the porcine cellular innate immunity during infection. The innate immune system of the porcine lung includes alveolar and parenchymal/interstitial macrophages, neutrophils (PMN), conventional dendritic cells (DC) and plasmacytoid DC, natural killer cells, and γδ T cells, thus the in vitro and in vivo interactions between those cells and PRRSV and SwIV are reviewed. Likewise, the few studies regarding PRRSV-SwIV co-infection are illustrated together with the different modulation mechanisms that are induced by the two viruses. Alterations in responses by natural killer (NK), PMN, or γδ T cells have not received much attention within the scientific community as their counterpart antigen-presenting cells and there are numerous gaps in the knowledge regarding the role of those cells in both infections. This review will help in paving the way for future directions in PRRSV and SwIV research and enhancing the understanding of the innate mechanisms that are involved during infection with these viruses.
Collapse
|
28
|
Ogno G, Sautter CA, Canelli E, García-Nicolás O, Stadejek T, Martelli P, Borghetti P, Summerfield A. In vitro characterization of PRRSV isolates with different in vivo virulence using monocyte-derived macrophages. Vet Microbiol 2019; 231:139-146. [PMID: 30955801 DOI: 10.1016/j.vetmic.2019.03.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 03/07/2019] [Accepted: 03/10/2019] [Indexed: 01/21/2023]
Abstract
The recent emergence of highly pathogenic porcine reproductive and respiratory syndrome virus 1 (PRRSV-1) strains has caused severe economic losses. The biological elements defining virulence and pathogenicity are still unclear. In vitro characteristics using natural target cells of PRRSV provide important information to understand the basis of virulence at the cellular level, and provide a mean to reduce animal experimentations to achieve this goal. Here, we compared PRRSV strains from two geographically different regions, with varying in vivo characteristics, in terms of their interactions with monocyte-derived macrophages (MDMs). The strains included Lena and BOR59 from Belarus, and ILI6 from Russia, as well as PR11 and PR40, both from Italy. As a reference, we used a cell culture-adapted version of Lelystad, LVP. MDMs were pre-treated with IFNγ, IL-4 or IFNβ, in order to understand responses in polarized and antiviral MDMs. In general, independent of the geographical origin, the strains with high virulence infected a higher percentage of MDMs and replicated to higher titers. These virulence-dependent differences were most pronounced when the MDMs had been treated with IFNβ. Differentiation between intermediate and low virulent PRRSV was difficult, due to variations between different experiments, but LVP differed clearly from all field strains. IFNα and IL-10 were not detected in any experiment, but PR40 induced TNF and IL-1β. Taken together, these results validate the MDM model to understand pathogenicity factors of PRRSV and confirm the importance of the escape from type I and II IFN-mediated effects for PRRSV virulence.
Collapse
Affiliation(s)
- Giulia Ogno
- Department of Veterinary Science, University of Parma, Strada del Taglio, Parma, 10 - 43126, Italy
| | - Carmen A Sautter
- Institute of Virology and Immunology, Bern, Mittelhäusern, 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
| | - Elena Canelli
- Department of Veterinary Science, University of Parma, Strada del Taglio, Parma, 10 - 43126, Italy
| | - Obdulio García-Nicolás
- Institute of Virology and Immunology, Bern, Mittelhäusern, Switzerland; Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Tomasz Stadejek
- Department of Pathology and Veterinary Diagnostics, Faculty of Veterinary Medicine, Warsaw University of Life Sciences, Poland
| | - Paolo Martelli
- Department of Veterinary Science, University of Parma, Strada del Taglio, Parma, 10 - 43126, Italy
| | - Paolo Borghetti
- Department of Veterinary Science, University of Parma, Strada del Taglio, Parma, 10 - 43126, Italy
| | - Artur Summerfield
- Institute of Virology and Immunology, Bern, Mittelhäusern, Switzerland; Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland.
| |
Collapse
|
29
|
Montaner-Tarbes S, Del Portillo HA, Montoya M, Fraile L. Key Gaps in the Knowledge of the Porcine Respiratory Reproductive Syndrome Virus (PRRSV). Front Vet Sci 2019; 6:38. [PMID: 30842948 PMCID: PMC6391865 DOI: 10.3389/fvets.2019.00038] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 01/30/2019] [Indexed: 12/11/2022] Open
Abstract
The porcine reproductive and respiratory syndrome virus (PRRSV) is one of the most important swine diseases in the world. It is causing an enormous economic burden due to reproductive failure in sows and a complex respiratory syndrome in pigs of all ages, with mortality varying from 2 to 100% in the most extreme cases of emergent highly pathogenic strains. PRRSV displays complex interactions with the immune system and a high mutation rate, making the development, and implementation of control strategies a major challenge. In this review, the biology of the virus will be addressed focusing on newly discovered functions of non-structural proteins and novel dissemination mechanisms. Secondly, the role of different cell types and viral proteins will be reviewed in natural and vaccine-induced immune response together with the role of different immune evasion mechanisms focusing on those gaps of knowledge that are critical to generate more efficacious vaccines. Finally, novel strategies for antigen discovery and vaccine development will be discussed, in particular the use of exosomes (extracellular vesicles of endocytic origin). As nanocarriers of lipids, proteins and nucleic acids, exosomes have potential effects on cell activation, modulation of immune responses and antigen presentation. Thus, representing a novel vaccination approach against this devastating disease.
Collapse
Affiliation(s)
- Sergio Montaner-Tarbes
- Innovex Therapeutics S.L, Badalona, Spain.,Departamento de Ciencia Animal, Escuela Técnica Superior de Ingenieria Agraria (ETSEA), Universidad de Lleida, Lleida, Spain
| | - Hernando A Del Portillo
- Innovex Therapeutics S.L, Badalona, Spain.,Germans Trias i Pujol Health Science Research Institute, Badalona, Spain.,ISGlobal, Hospital Clínic-Universitat de Barcelona, Barcelona, Spain.,Institució Catalana de Recerca i Estudis Avançats, Barcelona, Spain
| | - María Montoya
- Innovex Therapeutics S.L, Badalona, Spain.,Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Cientificas, Madrid, Spain
| | - Lorenzo Fraile
- Innovex Therapeutics S.L, Badalona, Spain.,Departamento de Ciencia Animal, Escuela Técnica Superior de Ingenieria Agraria (ETSEA), Universidad de Lleida, Lleida, Spain
| |
Collapse
|
30
|
Shabir N, Khatun A, Nazki S, Gu S, Lee SM, Hur TY, Yang MS, Kim B, Kim WI. In vitro immune responses of porcine alveolar macrophages reflect host immune responses against porcine reproductive and respiratory syndrome viruses. BMC Vet Res 2018; 14:380. [PMID: 30509265 PMCID: PMC6278023 DOI: 10.1186/s12917-018-1675-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 10/29/2018] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Currently, an in vitro immunogenicity screening system for the immunological assessment of potential porcine reproductive and respiratory syndrome virus (PRRSV) vaccine candidates is highly desired. Thus, in the present study, two genetically divergent PRRSVs were characterized in vitro and in vivo to identify an in vitro system and immunological markers that predict the host immune response. Porcine alveolar macrophages (PAMs) and peripheral blood mononuclear cells (PBMCs) collected from PRRSV-negative pigs were used for in vitro immunological evaluation, and the response of these cells to VR2332c or JA142c were compared with those elicited in pigs challenged with the same viruses. RESULTS Compared with VR2332c or mock infection, JA142c induced increased levels of type I interferons and pro-inflammatory cytokines (TNF-α, IL-1α/β, IL-6, IL-8, and IL-12) in PAMs, and these elevated levels were comparable to the cytokine induction observed in PRRSV-challenged pigs. Furthermore, significantly greater numbers of activated CD4+ T cells, type I helper T cells, cytotoxic T cells and total IFN-γ+ cells were observed in JA142c-challenged pigs than in VR2332c- or mock-challenged pigs. CONCLUSIONS Based on these results, the innate immune response patterns (particularly IFN-α, TNF-α and IL-12) to specific PRRSV strains in PAMs might reflect those elicited by the same viruses in pigs.
Collapse
Affiliation(s)
- Nadeem Shabir
- College of Veterinary Medicine, Chonbuk National University, 79 Gobong-ro, Iksan, Jeonbuk, Korea.,Division of Animal Biotechnology, Faculty of Veterinary Sciences and Animal Husbandry, Sher-e-Kashmir University of Agricultural Sciences and Technology of Kashmir, Srinagar, India
| | - Amina Khatun
- College of Veterinary Medicine, Chonbuk National University, 79 Gobong-ro, Iksan, Jeonbuk, Korea
| | - Salik Nazki
- College of Veterinary Medicine, Chonbuk National University, 79 Gobong-ro, Iksan, Jeonbuk, Korea
| | - Suna Gu
- College of Environmental & Biosource Science, Division of Biotechnology, Chonbuk National University, Iksan, South Korea
| | - Sang-Myoung Lee
- College of Environmental & Biosource Science, Division of Biotechnology, Chonbuk National University, Iksan, South Korea
| | - Tai-Young Hur
- Dairy Science Division, National Institute of Animal Science, Rural Development Administration, Cheonan, 31000, South Korea
| | - Myoun-Sik Yang
- College of Veterinary Medicine, Chonbuk National University, 79 Gobong-ro, Iksan, Jeonbuk, Korea
| | - Bumseok Kim
- College of Veterinary Medicine, Chonbuk National University, 79 Gobong-ro, Iksan, Jeonbuk, Korea
| | - Won-Il Kim
- College of Veterinary Medicine, Chonbuk National University, 79 Gobong-ro, Iksan, Jeonbuk, Korea.
| |
Collapse
|
31
|
Li YL, Darwich L, Mateu E. Characterization of the attachment and infection by Porcine reproductive and respiratory syndrome virus 1 isolates in bone marrow-derived dendritic cells. Vet Microbiol 2018; 223:181-188. [PMID: 30173745 DOI: 10.1016/j.vetmic.2018.08.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 08/09/2018] [Accepted: 08/10/2018] [Indexed: 01/11/2023]
Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) is known to infect porcine dendritic cells (DC). Previous studies indicated that different PRRSV1 isolates regulated differently the cytokine profiles and expression of surface molecules of DC. However, the characterisation of the infection is lacking. The current study aimed to characterise the replication and attachment of different PRRSV1 isolates in bone marrow-derived DC (BMDC). For this purpose, immature (i) and mature (m) BMDC were infected with three PRRSV1 isolates. The replication kinetics showed that titres in iBMDC were significantly (p < 0.05) higher than in mBMDC by 24 hpi, and for two isolates titres peaked earlier in iBMDC, suggesting that iBMDC were more efficient in supporting PRRSV1 replication than mBMDC. The attachment was revealed by a three-colour confocal microscopy staining. All three isolates were seen attached to iBMDC even in cells lacking CD163 -the essential receptor for PRRSV- or porcine sialoadhesin (PoSn). The attachment was not fully avoided after removal of heparan sulphate by heparinase I. Furthermore, the infection was examined with regards to CD163 expression. By flow cytometry and confocal microscopy, positive signals of PRRSV1 nucleocapsid could be observed in CD163- iBMDC. Additional sorting experiment demonstrated that CD163- iBMDC were infected only when CD163lo/hi cells were present. This can be interpreted in different ways: susceptible CD163- cells arose as result of milieu created by CD163+ infected BMDC; CD163- cells were infected by receptor-independent mechanisms (i.e. exosomes) or, some cells expressed CD163 at levels beyond the technical sensitivity.
Collapse
Affiliation(s)
- Yan-Li Li
- Departament de Sanitat i Anatomia Animals, Universitat Autònoma de Barcelona, 08193 Cerdanyola Del Vallès, Spain; IRTA, Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Campus de la Universitat Autònoma de Barcelona, 08193 Cerdanyola Del Vallès, Spain.
| | - Laila Darwich
- Departament de Sanitat i Anatomia Animals, Universitat Autònoma de Barcelona, 08193 Cerdanyola Del Vallès, Spain; IRTA, Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Campus de la Universitat Autònoma de Barcelona, 08193 Cerdanyola Del Vallès, Spain
| | - Enric Mateu
- Departament de Sanitat i Anatomia Animals, Universitat Autònoma de Barcelona, 08193 Cerdanyola Del Vallès, Spain; IRTA, Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Campus de la Universitat Autònoma de Barcelona, 08193 Cerdanyola Del Vallès, Spain
| |
Collapse
|
32
|
Fernández-Caballero T, Álvarez B, Alonso F, Revilla C, Martínez-Lobo J, Prieto C, Ezquerra Á, Domínguez J. Interaction of PRRS virus with bone marrow monocyte subsets. Vet Microbiol 2018; 219:123-127. [PMID: 29778184 DOI: 10.1016/j.vetmic.2018.04.027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 04/16/2018] [Accepted: 04/16/2018] [Indexed: 11/29/2022]
Abstract
PRRSV can replicate for months in lymphoid organs leading to persistent host infections. Porcine bone marrow comprises two major monocyte subsets, one of which expresses CD163 and CD169, two receptors involved in the entry of PRRSV in macrophages. In this study, we investigate the permissiveness of these subsets to PRRSV infection. PRRSV replicates efficiently in BM CD163+ monocytes reaching titers similar to those obtained in alveolar macrophages, but with a delayed kinetics. Infection of BM CD163- monocytes was variable and yielded lower titers. This may be related with the capacity of BM CD163- monocytes to differentiate into CD163+ CD169+ cells after culture in presence of M-CSF. Both subsets secreted IL-8 in response to virus but CD163+ cells tended to produce higher amounts. The infection of BM monocytes by PRRSV may contribute to persistence of the virus in this compartment and to hematological disorders found in infected animals such as the reduction in the number of peripheral blood monocytes.
Collapse
Affiliation(s)
- Teresa Fernández-Caballero
- Dpto. Biotecnología, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), 28040, Madrid, Spain
| | - Belén Álvarez
- Dpto. Biotecnología, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), 28040, Madrid, Spain
| | - Fernando Alonso
- Dpto. Biotecnología, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), 28040, Madrid, Spain
| | - Concepción Revilla
- Dpto. Biotecnología, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), 28040, Madrid, Spain
| | - Javier Martínez-Lobo
- Dpto. Sanidad Animal. Facultad de Veterinaria, Universidad Complutense de Madrid (UCM), 28040, Madrid, Spain
| | - Cinta Prieto
- Dpto. Sanidad Animal. Facultad de Veterinaria, Universidad Complutense de Madrid (UCM), 28040, Madrid, Spain
| | - Ángel Ezquerra
- Dpto. Biotecnología, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), 28040, Madrid, Spain
| | - Javier Domínguez
- Dpto. Biotecnología, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), 28040, Madrid, Spain.
| |
Collapse
|
33
|
Comparative evaluation of immune responses of swine in PRRS-stable and unstable herds. Vet Immunol Immunopathol 2018; 200:32-39. [PMID: 29776610 DOI: 10.1016/j.vetimm.2018.04.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 04/11/2018] [Accepted: 04/15/2018] [Indexed: 11/24/2022]
Abstract
Porcine Reproductive and Respiratory Syndrome (PRRS) is an elusive model of host/virus relationship in which disease is determined by virus pathogenicity, pig breed susceptibility and phenotype, microbial infectious pressure and environmental conditions. Successful disease control in PRRS-endemic Countries corresponds to "stability", i.e. a condition with no clinical signs of PRRS in the breeding-herd population and no viremia in weaning-age pigs. The aim of this work was to compare the profile and time-course of humoral and cell-mediated immunity in stable and unstable herds, respectively. In particular, we investigated PRRS virus (PRRSV) in serum and group oral fluid samples by Real-time RT-PCR, PRRSV-specific IgA and IgG in oral fluids, serum IgG antibody and the cell-mediated response (PRRSV-specific release of interferon-gamma) in whole blood samples. These parameters were measured in order to identify possible discrepancies in the development and kinetics of the immune response against PRRSV. PRRS-free gilts got regularly infected after entering PRRS-stable and unstable farms. In an open cycle, unstable pig farm PRRSV infection could be demonstrated in all groups of pigs, including suckling piglets. Four main results should be highlighted: A) the precocity of the Ab response in group oral fluids was generally similar to that recorded in sera; B) circulation of PRRSV was consistently detected in all age groups in the unstable herds, as opposed to the stable ones; C) an early, balanced, IgA and IgG response in oral fluids was only observed in the stable herds; D) an early IFN-gamma response after PRRSV infection was often observed in stable herds, as opposed to the unstable ones. These were characterized by IFN-gamma responses in piglets, likely due to transfer of maternal immunity. Most important, the mucosal IgA response was associated with cessation of virus excretion in oral fluid samples of PRRS-unstable herds. The above findings indicate that a peculiar profile of immune response to PRRSV can be found in PRRS-stable herds. Therefore, the outlined immune parameters can represent a useful readout system to evaluate successful adaptation to PRRSV based on acclimatization of breeding animals and management of pig flow.
Collapse
|
34
|
An CH, Nazki S, Park SC, Jeong YJ, Lee JH, Park SJ, Khatun A, Kim WI, Park YI, Jeong JC, Kim CY. Plant synthetic GP4 and GP5 proteins from porcine reproductive and respiratory syndrome virus elicit immune responses in pigs. PLANTA 2018; 247:973-985. [PMID: 29313103 DOI: 10.1007/s00425-017-2836-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 12/13/2017] [Indexed: 06/07/2023]
Abstract
MAIN CONCLUSION We demonstrated successful overexpression of porcine reproductive and respiratory syndrome virus (PRRSV)-derived GP4D and GP5D antigenic proteins in Arabidopsis. Pigs immunized with transgenic plants expressing GP4D and GP5D proteins generated both humoral and cellular immune responses to PRRSV. Porcine reproductive and respiratory syndrome virus (PRRSV) causes PRRS, the most economically significant disease affecting the swine industry worldwide. However, current commercial PRRSV vaccines (killed virus or modified live vaccines) show poor efficacy and safety due to concerns such as reversion of virus to wild type and lack of cross protection. To overcome these problems, plants are considered a promising alternative to conventional platforms and as a vehicle for large-scale production of recombinant proteins. Here, we demonstrate successful production of recombinant protein vaccine by expressing codon-optimized and transmembrane-deleted recombinant glycoproteins (GP4D and GP5D) from PRRSV in planta. We generated transgenic Arabidopsis plants expressing GP4D and GP5D proteins as candidate antigens. To examine immunogenicity, pigs were fed transgenic Arabidopsis leaves expressing the GP4D and GP5D antigens (three times at 2-week intervals) and then challenged with PRRSV at 6-week post-initial treatment. Immunized pigs showed significantly lower lung lesion scores and reduced viremia and viral loads in the lung than pigs fed Arabidopsis leaves expressing mYFP (control). Immunized pigs also had higher titers of PRRSV-specific antibodies and significantly higher levels of pro-inflammatory cytokines (TNF-α and IL-12). Furthermore, the numbers of IFN-γ+-producing cells were higher, and those of regulatory T cells were lower, in GP4D and GP5D immunized pigs than in control pigs. Thus, plant-derived GP4D and GP5D proteins provide an alternative platform for producing an effective subunit vaccine against PRRSV.
Collapse
MESH Headings
- Animals
- Antibodies, Viral/immunology
- Antigens, Viral/immunology
- Arabidopsis/genetics
- Arabidopsis/metabolism
- Blotting, Western
- Enzyme-Linked Immunosorbent Assay
- Flow Cytometry
- Immunity, Cellular
- Immunity, Humoral
- Leukocytes, Mononuclear/immunology
- Plants, Genetically Modified/genetics
- Plants, Genetically Modified/metabolism
- Porcine respiratory and reproductive syndrome virus/immunology
- Reverse Transcriptase Polymerase Chain Reaction
- Swine/immunology
- Swine/virology
- Vaccines, Synthetic/biosynthesis
- Vaccines, Synthetic/immunology
Collapse
Affiliation(s)
- Chul Han An
- Biological Resource Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 181 Ipsin-gil, Jeongeup, Jeonbuk, 56212, Republic of Korea
- Department of Bioscience and Biotechnology, Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Salik Nazki
- College of Veterinary Medicine and College of Environmental and Biosource Science, Chonbuk National University, Iksan, Jeonbuk, 54596, Republic of Korea
| | - Sung-Chul Park
- Biological Resource Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 181 Ipsin-gil, Jeongeup, Jeonbuk, 56212, Republic of Korea
| | - Yu Jeong Jeong
- Biological Resource Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 181 Ipsin-gil, Jeongeup, Jeonbuk, 56212, Republic of Korea
| | - Ju Huck Lee
- Biological Resource Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 181 Ipsin-gil, Jeongeup, Jeonbuk, 56212, Republic of Korea
| | - Su-Jin Park
- Natural Product Material Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 181 Ipsin-gil, Jeongeup, Jeonbuk, 56212, Republic of Korea
| | - Amina Khatun
- College of Veterinary Medicine and College of Environmental and Biosource Science, Chonbuk National University, Iksan, Jeonbuk, 54596, Republic of Korea
| | - Won-Il Kim
- College of Veterinary Medicine and College of Environmental and Biosource Science, Chonbuk National University, Iksan, Jeonbuk, 54596, Republic of Korea
| | - Youn-Il Park
- Department of Bioscience and Biotechnology, Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Jae Cheol Jeong
- Biological Resource Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 181 Ipsin-gil, Jeongeup, Jeonbuk, 56212, Republic of Korea.
| | - Cha Young Kim
- Biological Resource Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 181 Ipsin-gil, Jeongeup, Jeonbuk, 56212, Republic of Korea.
| |
Collapse
|
35
|
Canelli E, Catella A, Borghetti P, Ferrari L, Ogno G, De Angelis E, Corradi A, Passeri B, Bertani V, Sandri G, Bonilauri P, Leung FC, Guazzetti S, Martelli P. Phenotypic characterization of a highly pathogenic Italian porcine reproductive and respiratory syndrome virus (PRRSV) type 1 subtype 1 isolate in experimentally infected pigs. Vet Microbiol 2017; 210:124-133. [DOI: 10.1016/j.vetmic.2017.09.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 09/01/2017] [Accepted: 09/06/2017] [Indexed: 12/11/2022]
|
36
|
Cortey M, Díaz I, Martín-Valls G, Mateu E. Next-generation sequencing as a tool for the study of Porcine reproductive and respiratory syndrome virus (PRRSV) macro- and micro- molecular epidemiology. Vet Microbiol 2017; 209:5-12. [DOI: 10.1016/j.vetmic.2017.02.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 01/31/2017] [Accepted: 02/02/2017] [Indexed: 12/20/2022]
|
37
|
Gibert E, Martín-Valls G, Mateu E. Comparison of protocols for the analysis of type 1 porcine reproductive and respiratory syndrome virus by RT-PCR using oral fluids. J Virol Methods 2017; 243:190-195. [PMID: 28213086 DOI: 10.1016/j.jviromet.2017.02.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 01/12/2017] [Accepted: 02/13/2017] [Indexed: 11/17/2022]
Abstract
The detection of porcine reproductive and respiratory syndrome virus (PRRSV) in oral fluids (OF) by quantitative real-time polymerase chain reaction (qRT-PCR) is gaining increasing popularity. However, the different steps leading to a result have not been extensively evaluated. The aim of the present study was to examine the effect on the performance of qRT-PCR with different sampling materials, conditions of storage of the OF, the need for centrifuging OF, as well as to compare RNA extraction methods and PCR mixes. For the assays, pen-based oral fluids were used, which were pooled and spiked in a serial dilution (up to genotype 100 TCID50/mL) of type 1 PRRSV isolate 3267. Centrifugation at 15,000g for 15min resulted in an increase in sensitivity (1-2 PCR cycles) that was significant (P<0.05) at the lowest dilution tested. The TRIzol and MagMAX RNA extraction methods gave the maximum sensitivity, lowest threshold cycle (Ct), at equivalent virus concentrations. The AgPath-ID One-Step RT-PCR Kit PCR mix reagents were more sensitive for the detection of PRRSV using a purified plasmid as standard, but LSI VetMAX PRRSV EU/NA PRRSV reagents resulted in a slightly better sensitivity with OF (p<0.05). The present results may be useful to standardize protocols for optimizing detection of type 1 PRRSV in OF by qRT-PCR.
Collapse
Affiliation(s)
- Elisa Gibert
- Centre de Recerca en Sanitat Animal (CReSA), IRTA-UAB, Edifici CReSA, Campus UAB, 08193, Bellaterra, Spain.
| | - Gerard Martín-Valls
- Departament de Sanitat i Anatomia Animals, Universitat Autònoma de Barcelona (UAB), Campus UAB, 08193, Bellaterra, Spain.
| | - Enric Mateu
- Centre de Recerca en Sanitat Animal (CReSA), IRTA-UAB, Edifici CReSA, Campus UAB, 08193, Bellaterra, Spain; Departament de Sanitat i Anatomia Animals, Universitat Autònoma de Barcelona (UAB), Campus UAB, 08193, Bellaterra, Spain.
| |
Collapse
|
38
|
Nedumpun T, Wongyanin P, Sirisereewan C, Ritprajak P, Palaga T, Thanawongnuwech R, Suradhat S. Interleukin-1 receptor antagonist: an early immunomodulatory cytokine induced by porcine reproductive and respiratory syndrome virus. J Gen Virol 2017; 98:77-88. [PMID: 27902420 DOI: 10.1099/jgv.0.000665] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) infection poorly induces pro-inflammatory cytokines (IL-1, IL-6 and TNF-α) and type I IFN production during the early phase of infection. Our microarray analysis indicated strong upregulation of the IL1RA gene in type 2 PRRSV -infected monocyte-derived dendritic cells. Interleukin-1 receptor antagonist (IL-1Ra) is an early inhibitory cytokine that suppresses pro-inflammatory cytokines and T-lymphocyte responses. To investigate the induction of IL-1Ra by PRRSV, monocyte-derived dendritic cells were cultured with type 2 PRRSV or other swine viruses. PRRSV increased both IL1RA gene expression and IL-1Ra protein production in the culture. The enhanced production of IL-1Ra was further confirmed in PRRSV-cultured PBMC and PRRSV-exposed pigs by flow cytometry. Myeloid cell population appeared to be the major IL-1Ra producer both in vitro and in vivo. In contrast to the type 2 PRRSV, the highly pathogenic (HP)- PRRSV did not upregulate IL1RA gene expression in vitro. To determine the kinetics of PRRSV-induced IL1RA gene expression in relation to other pro-inflammatory cytokine genes, PRRSV-negative pigs were vaccinated with a commercially available type 2 modified-live PRRS vaccine or intranasally inoculated with HP-PRRSV. In modified-live PRRS vaccine pigs, upregulation of IL1RA, but not IL1B and IFNA, gene expression was observed from 2 days post- vaccination. Consistent with the in vitro findings, upregulation of IL1RA gene expression was not observed in the HP-PRRSV-infected pigs throughout the experiment. This study identified IL-1Ra as an early immunomodulatory mediator that could be involved in the immunopathogenesis of PRRSV infections.
Collapse
Affiliation(s)
- Teerawut Nedumpun
- Interdisciplinary Program of Medical Microbiology, Graduate School, Chulalongkorn University, Bangkok, Thailand
| | - Piya Wongyanin
- Department of Medical Technology, Faculty of Science and Technology, Bansomdejchaopraya Rajabhat University, Bangkok, Thailand
| | - Chaitawat Sirisereewan
- Graduate Program in Veterinary Pathobiology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
| | - Patcharee Ritprajak
- Department of Microbiology, RU in Oral Microbiology and Immunology, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Tanapat Palaga
- Department of Microbiology, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
| | - Roongroje Thanawongnuwech
- Center of Excellence in Emerging Infectious Diseases in Animals, Chulalongkorn University (CU-EIDAs), Bangkok, Thailand.,Department of Veterinary Pathology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
| | - Sanipa Suradhat
- Department of Veterinary Microbiology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand.,Center of Excellence in Emerging Infectious Diseases in Animals, Chulalongkorn University (CU-EIDAs), Bangkok, Thailand
| |
Collapse
|
39
|
Pileri E, Martín-Valls GE, Díaz I, Allepuz A, Simon-Grifé M, García-Saenz A, Casal J, Mateu E. Estimation of the transmission parameters for swine influenza and porcine reproductive and respiratory syndrome viruses in pigs from weaning to slaughter under natural conditions. Prev Vet Med 2017; 138:147-155. [PMID: 28237230 DOI: 10.1016/j.prevetmed.2017.01.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 01/10/2017] [Accepted: 01/15/2017] [Indexed: 01/16/2023]
Abstract
In the present study, the transmission parameters of swine influenza virus (SIV) and porcine reproductive and respiratory virus (PRRSV) have been calculated using the basic reproductive rate (R) parameter in two commercial pig farms (F1 and F2). In order to do this, a serological (PRRSV genotype 1 and SIV) and virological (SIV) follow-up of a batch of animals was carried out weekly from 3 weeks of age until the age of slaughter on each farm. Results of the analysis for SIV and PRRSV showed different transmission profiles depending on the farm, the pathogen, and time of transmission. In F1, transmission of both viruses was detected throughout the sampling. The Rt (R for a given period of time) value for SIV ranged from 1.5 [0.9-2.3] to 3.6 [2.3-4.9] from farrowing to the beginning of the fattening period, and the Rt value for PRRSV was 3.3 [2.9-4.3] to 3.5 [2.8-4.1] from farrowing until the slaughter age. These results indicated that both viruses were transmitted enzootically in that farm for these periods of time. A different transmission pattern with a higher incidence was also observed during the fattening period in F1 (after 15 weeks of age) for SIV, coinciding with the entrance of a new subtype. In this case, R value for SIV reached 3.3 [1.65-4.9]. On the other hand, in F2, SIV and PRRSV seemed to be restricted to the fattening period. R reached a value of 6.4 [4.1-8.8] for SIV and 7.1 [3.5-10.6] for PRRSV. These findings suggest a different origin of the virus, as well as a more epidemic circulation, especially for SIV, where most of the new cases were observed in a one week period. In conclusion, the present study offers a reliable estimation of the range of Rt values for SIV and genotype 1 PRRSV transmission under field conditions, suggesting that enzootic circulations of both viruses are similar in terms of transmission, probably higher for PRRSV, but also that transmission of SIV is more efficient (or epidemic) than transmission of a genotype 1 PRRSV isolate in naïve animals given the new cases observed in only in F2.
Collapse
Affiliation(s)
- Emanuela Pileri
- IRTA, Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Campus de la Universitat Autònoma de Barcelona, 08193 Bellaterra, Barcelona, Spain,; Departament de Sanitat i d'Anatomia Animals, Universitat Autònoma de Barcelona, 08193 Bellaterra (Cerdanyola del Vallès), Spain
| | - Gerard E Martín-Valls
- IRTA, Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Campus de la Universitat Autònoma de Barcelona, 08193 Bellaterra, Barcelona, Spain,; Departament de Sanitat i d'Anatomia Animals, Universitat Autònoma de Barcelona, 08193 Bellaterra (Cerdanyola del Vallès), Spain.
| | - Ivan Díaz
- IRTA, Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Campus de la Universitat Autònoma de Barcelona, 08193 Bellaterra, Barcelona, Spain
| | - Alberto Allepuz
- IRTA, Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Campus de la Universitat Autònoma de Barcelona, 08193 Bellaterra, Barcelona, Spain,; Departament de Sanitat i d'Anatomia Animals, Universitat Autònoma de Barcelona, 08193 Bellaterra (Cerdanyola del Vallès), Spain.
| | - Meritxell Simon-Grifé
- IRTA, Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Campus de la Universitat Autònoma de Barcelona, 08193 Bellaterra, Barcelona, Spain
| | - Ariadna García-Saenz
- IRTA, Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Campus de la Universitat Autònoma de Barcelona, 08193 Bellaterra, Barcelona, Spain,; Departament de Sanitat i d'Anatomia Animals, Universitat Autònoma de Barcelona, 08193 Bellaterra (Cerdanyola del Vallès), Spain
| | - Jordi Casal
- IRTA, Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Campus de la Universitat Autònoma de Barcelona, 08193 Bellaterra, Barcelona, Spain,; Departament de Sanitat i d'Anatomia Animals, Universitat Autònoma de Barcelona, 08193 Bellaterra (Cerdanyola del Vallès), Spain
| | - Enric Mateu
- IRTA, Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Campus de la Universitat Autònoma de Barcelona, 08193 Bellaterra, Barcelona, Spain,; Departament de Sanitat i d'Anatomia Animals, Universitat Autònoma de Barcelona, 08193 Bellaterra (Cerdanyola del Vallès), Spain
| |
Collapse
|
40
|
Diseases Primarily Affecting the Reproductive System. Vet Med (Auckl) 2017. [PMCID: PMC7150237 DOI: 10.1016/b978-0-7020-5246-0.00018-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
41
|
Transmission of Porcine reproductive and respiratory syndrome virus 1 to and from vaccinated pigs in a one-to-one model. Vet Microbiol 2016; 201:18-25. [PMID: 28284607 DOI: 10.1016/j.vetmic.2016.12.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 12/09/2016] [Accepted: 12/09/2016] [Indexed: 11/23/2022]
Abstract
The present study examined transmission by contact of Porcine reproductive and respiratory syndrome virus (PRRSV) 1 in a one-to-one model to vaccinated and unvaccinated pigs and from vaccinated infected pigs to other vaccinated pigs. The experiment started by randomly assigning weaned pigs to groups V (n=24) and U (n=26). V pigs were vaccinated with a commercial live attenuated PRRSV vaccine and the U animals were kept as unvaccinated controls. Twenty-eight days later, 6U pigs were separated and allocated in individual boxes. The remaining 20U pigs were intranasally inoculated with PRRSV isolate 3267 (from now on designated as seeder (S) pigs) and 48h later were distributed in boxes where they were commingled with either V or U pigs in 1:1 groups (first contact phase), resulting in 6S:U and 14S:V pairs. As soon as a V pig was detected to be viremic because of contact with a S, the infected V (from now on designated as Vinf) was transferred (<24h after detection) to a new pen where it was comingled with a new V pig (designated as V2) in a second contact phase. For the first contact phase, pigs were maintained 21days at maximum and for the second contact phase the maximum exposure period was 14days. Two V pigs tested positive for the vaccine virus (>99.5% similarity) when they were relocated with the corresponding V2 pigs and they were removed; thus, only 12Vinf were finally considered. All V pigs (12/12) exposed to S animals became infected although the first detection of viremia occurred at 13.6±3.6days, one week later than in U (p<0.05). Also, duration of viremia was shorter for Vinf compared to U, (5.5±4.3days versus 12.5±2.7days). The Vinf group showed remarkable individual variability: eight animals had a viremic period of 5 or less days (3.0±1.4) while the remaining four had a longer viremic period of more than one week (10.8±2.9). This situation was not observed in U. In the second contact phase, transmission from Vinf to V2 pigs occurred in 7/8 cases (87.5%). The mean duration of viremia for V2 was 4.8±3.4 and two different patterns were again observed: two animals had viremias of 9-10days and the rest averaged 3.0±1.4days (range: 2-5days). Vaccinated groups Vinf and V2 had a significantly lower PRRSV shedding in oral fluids for at least the first 9days after the onset of the viremia compared to U, and shedding for V2 was even significantly lower (p<0.05) than shedding for Vinf. Our experimental design reproduced the worst-case scenario for evaluating the effect of vaccination and, under such conditions; it was still efficacious in slowering PRRSV transmission and decreasing the global viral load and particularly oral shedding.
Collapse
|
42
|
Doeschl-Wilson A, Wilson A, Nielsen J, Nauwynck H, Archibald A, Ait-Ali T. Combining laboratory and mathematical models to infer mechanisms underlying kinetic changes in macrophage susceptibility to an RNA virus. BMC SYSTEMS BIOLOGY 2016; 10:101. [PMID: 27770812 PMCID: PMC5075420 DOI: 10.1186/s12918-016-0345-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 10/14/2016] [Indexed: 01/04/2023]
Abstract
Background Macrophages are essential to innate immunity against many pathogens, but some pathogens also target macrophages as routes to infection. The Porcine Reproductive and Respiratory Syndrome virus (PRRSV) is an RNA virus that infects porcine alveolar macrophages (PAMs) causing devastating impact on global pig production. Identifying the cellular mechanisms that mediate PAM susceptibility to the virus is crucial for developing effective interventions. Previous evidence suggests that the scavenger receptor CD163 is essential for productive infection of PAMs with PRRSV. Here we use an integrative in-vitro–in-silico modelling approach to determine whether and how PAM susceptibility to PRRSV changes over time, to assess the role of CD163 expression on such changes, and to infer other potential causative mechanisms altering cell susceptibility. Results Our in-vitro experiment showed that PAM susceptibility to PRRSV changed considerably over incubation time. Moreover, an increasing proportion of PAMs apparently lacking CD163 were found susceptible to PRRSV at the later incubation stages, thus conflicting with current understanding that CD163 is essential for productive infection of PAMs with PRRSV. We developed process based dynamic mathematical models and fitted these to the data to assess alternative hypotheses regarding potential underlying mechanisms for the observed susceptibility and biomarker trends. The models informed by our data support the hypothesis that although CD163 may have enhanced cell susceptibility, it was not essential for productive infection in our study. Instead the models promote the existence of a reversible cellular state, such as macrophage polarization, mediated in a density dependent manner by autocrine factors, to be responsible for the observed kinetics in cell susceptibility. Conclusions Our dynamic model–inference approach provides strong support that PAM susceptibility to the PRRS virus is transient, reversible and can be mediated by compounds produced by the target cells themselves, and that these can render PAMs lacking the CD163 receptor susceptible to PRRSV. The results have implications for the development of therapeutics aiming to boost target cell resistance and prompt future investigation of dynamic changes in macrophage susceptibility to PRRSV and other viruses. Electronic supplementary material The online version of this article (doi:10.1186/s12918-016-0345-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Andrea Doeschl-Wilson
- The Roslin Institute & R(D)SVS, University of Edinburgh, Easter Bush, Edinburgh, UK.
| | - Alison Wilson
- The Roslin Institute & R(D)SVS, University of Edinburgh, Easter Bush, Edinburgh, UK
| | - Jens Nielsen
- Department of Mircrobiological Diagnostics and Virology, Statens Serum Institute, Copenhagen, Denmark
| | - Hans Nauwynck
- Department of Virology, Parasitology and Immunity, Ghent University, Ghent, Belgium
| | - Alan Archibald
- The Roslin Institute & R(D)SVS, University of Edinburgh, Easter Bush, Edinburgh, UK
| | - Tahar Ait-Ali
- The Roslin Institute & R(D)SVS, University of Edinburgh, Easter Bush, Edinburgh, UK
| |
Collapse
|
43
|
Ait-Ali T, Díaz I, Soldevila F, Cano E, Li Y, Wilson AD, Giotti B, Archibald AL, Mateu E, Darwich L. Distinct functional enrichment of transcriptional signatures in pigs with high and low IFN-gamma responses after vaccination with a porcine reproductive and respiratory syndrome virus (PRRSV). Vet Res 2016; 47:104. [PMID: 27765052 PMCID: PMC5073823 DOI: 10.1186/s13567-016-0392-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 10/05/2016] [Indexed: 01/05/2023] Open
Abstract
Little is known about the host factor in the response to PRRSV vaccination. For this purpose, piglets were immunized with a commercial PRRSV-live vaccine and classified as high responders (HR) or low responders (LR) as regards to the frequencies of virus-specific IFN-γ-secreting cells. Six weeks post vaccination, PBMCs isolated from three individuals with the most extreme responses in each HR and LR groups and 3 unvaccinated controls, were either stimulated with phytohaemagglutinin, challenged with the vaccine or mock treated for 24 h, prior conducting transcriptional studies, gene ontology and pathway analyses. The LR group had very low neutralizing antibody levels and showed a higher number of down-regulated transcripts compared with the HR group (FDR < 0.2, P < 0.001). Down-regulated genes encoded chemoattractants, proinflammatory cytokines and the interferon-inducible GBP family, and showed enrichment in wounding (FDR < 3.6E-13), inflammation (FDR < 8E-12), defence (FDR < 8.7E-09) and immunity (FDR < 7.6E-08), suggesting immune response impairment. In the HR group, down-regulated genes were involved in protein transport (FDR < 4.77E-03), locomotory behavior (FDR < 5.47E-3), regulation of protein localization (FDR < 1.02E-02), and regulation of TNF superfamily member 15 and miR181. In contrast, the HR group presented up-regulated transcripts associated with wounding (FDR < 4.95). Moreover, IFN-γ was predicted to be an inhibited upstream regulator since IFN-γ pathways were associated with higher number of down-regulated genes in the LR (n = 40) than the HR (n = 10). Divergent responses to PRRSV-vaccination may be the result of the genetic background of the host.
Collapse
Affiliation(s)
- Tahar Ait-Ali
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, EH25 9RG, UK
| | - Ivan Díaz
- Centre de Recerca en Sanitat Animal (CReSA)- IRTA, Campus de la Universitat Autònoma de Barcelona (UAB), 08193, Cerdanyola del Valles, Spain
| | - Ferran Soldevila
- Virology Department, Animal and Plant Health and Agency, Addlestone, KT15 3NB, UK.,Department of Pathology and Pathogen Biology, Royal Veterinary College, Hatfield, AL9 7TA, UK
| | - Esmeralda Cano
- Centre de Recerca en Sanitat Animal (CReSA)- IRTA, Campus de la Universitat Autònoma de Barcelona (UAB), 08193, Cerdanyola del Valles, Spain
| | - Yanli Li
- Department Sanitat i Anatomia Animals, Faculty of Veterinary, UAB, 08193, Cerdanyola del Valles, Spain
| | - Alison D Wilson
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, EH25 9RG, UK
| | - Bruno Giotti
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, EH25 9RG, UK
| | - Alan L Archibald
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, EH25 9RG, UK
| | - Enric Mateu
- Centre de Recerca en Sanitat Animal (CReSA)- IRTA, Campus de la Universitat Autònoma de Barcelona (UAB), 08193, Cerdanyola del Valles, Spain.,Department Sanitat i Anatomia Animals, Faculty of Veterinary, UAB, 08193, Cerdanyola del Valles, Spain
| | - Laila Darwich
- Centre de Recerca en Sanitat Animal (CReSA)- IRTA, Campus de la Universitat Autònoma de Barcelona (UAB), 08193, Cerdanyola del Valles, Spain. .,Department Sanitat i Anatomia Animals, Faculty of Veterinary, UAB, 08193, Cerdanyola del Valles, Spain.
| |
Collapse
|
44
|
Evaluation of the Cross-Protective Efficacy of a Chimeric Porcine Reproductive and Respiratory Syndrome Virus Constructed Based on Two Field Strains. Viruses 2016; 8:v8080240. [PMID: 27556483 PMCID: PMC4997602 DOI: 10.3390/v8080240] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2016] [Revised: 07/27/2016] [Accepted: 08/16/2016] [Indexed: 11/17/2022] Open
Abstract
One of the major hurdles to porcine reproductive and respiratory syndrome (PRRS) vaccinology is the limited or no cross-protection conferred by current vaccines. To overcome this challenge, a PRRS chimeric virus (CV) was constructed using an FL12-based cDNA infectious clone in which open reading frames (ORFs) 3-4 and ORFs 5-6 were replaced with the two Korean field isolates K08-1054 and K07-2273,respectively. This virus was evaluated as a vaccine candidate to provide simultaneous protection against two genetically distinct PRRS virus (PRRSV) strains. Thirty PRRS-negative three-week-old pigs were divided into five groups and vaccinated with CV, K08-1054, K07-2273, VR-2332, or a mock inoculum. At 25 days post-vaccination (dpv), the pigs in each group were divided further into two groups and challenged with either K08-1054 or K07-2273. All of the pigs were observed until 42 dpv and were euthanized for pathological evaluation. Overall, the CV-vaccinated group exhibited higher levels of tumor necrosis factor-alpha (TNF-α), interferon-gamma (IFN-γ), and interleukin-12 (IL-12) expression and of serum virus-neutralizing antibodies compared with the other groups after vaccination and also demonstrated better protection levels against both viruses compared with the challenge control group. Based on these results, it was concluded that CV might be an effective vaccine model that can confer a broader range of cross-protection to various PRRSV strains.
Collapse
|
45
|
Ligation of porcine Fc gamma receptor III inhibits levels of antiviral cytokine in response to PRRSV infection in vitro. Res Vet Sci 2016; 105:47-52. [DOI: 10.1016/j.rvsc.2016.01.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Revised: 01/06/2016] [Accepted: 01/13/2016] [Indexed: 11/23/2022]
|
46
|
Gu W, Wang Y, Luo X, Feng L, Niu J, Tian Z, Li R, Xu Y, Guo L, Zhang J. Elevated plasma-soluble CD16 levels in porcine reproductive and respiratory syndrome virus-infected pigs: correlation with ADAM17-mediated shedding. J Gen Virol 2016; 97:632-638. [DOI: 10.1099/jgv.0.000368] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
|
47
|
Differences of immune responses between Tongcheng (Chinese local breed) and Large White pigs after artificial infection with highly pathogenic porcine reproductive and respiratory syndrome virus. Virus Res 2016; 215:84-93. [PMID: 26878768 DOI: 10.1016/j.virusres.2016.02.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Revised: 02/05/2016] [Accepted: 02/10/2016] [Indexed: 12/22/2022]
Abstract
Porcine reproductive and respiratory syndrome (PRRS) is one of the severest infectious diseases of pigs throughout the world. Pigs of different breeds infected with PRRS virus (PRRSV) have been reported to vary in their immune responses. Here, the differences of immune responses to highly pathogenic porcine reproductive and respiratory syndrome virus (HP-PRRSV) were investigated by artificially infecting Tongcheng (TC) pigs (a Chinese indigenous breed) and Large White (LW) pigs with PRRSV WUH3. Compared to LW pigs, TC pigs showed less severe symptoms and lower level of viral load. The routine blood test results indicated that TC pigs were relatively steady in terms of erythrocyte, leukocyte and platelet. Additionally, PRRSV infection induced higher IFN-γ activity in TC pigs, but stimulated an excessive level of IL-10 and IL-12p40 in LW pigs. Our study provides direct evidence that TC pigs have stronger resistance to early PRRSV infection than LW pigs, suggesting that the resistance of pigs to PRRSV is likely associated with breed differences.
Collapse
|
48
|
Production of porcine TNFα by ADAM17-mediated cleavage negatively regulates porcine reproductive and respiratory syndrome virus infection. Immunol Res 2016; 64:711-20. [DOI: 10.1007/s12026-015-8772-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
49
|
Fan B, Liu X, Bai J, Li Y, Zhang Q, Jiang P. The 15N and 46R Residues of Highly Pathogenic Porcine Reproductive and Respiratory Syndrome Virus Nucleocapsid Protein Enhance Regulatory T Lymphocytes Proliferation. PLoS One 2015; 10:e0138772. [PMID: 26397116 PMCID: PMC4580451 DOI: 10.1371/journal.pone.0138772] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 08/09/2015] [Indexed: 12/14/2022] Open
Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) negatively modulates host immune responses, resulting in persistent infection and immunosuppression. PRRSV infection increases the number of PRRSV-specific regulatory T lymphocytes (Tregs) in infected pigs. However, the target antigens for Tregs proliferation in PRRSV infection have not been fully understood. In this study, we demonstrated that the highly pathogenic PRRSV (HP-PRRSV) induced more CD4+CD25+Foxp3+ Tregs than classical PRRSV (C-PRRSV) strain. Of the recombinant GP5, M and N proteins of HP-PRRSV expressed in baculovirus expression systems, only N protein induced Tregs proliferation. The Tregs assays showed that three amino-acid regions, 15–21, 42–48 and 88–94, in N protein played an important role in induction of Tregs proliferation with synthetic peptides covering the whole length of N protein. By using reverse genetic methods, it was firstly found that the 15N and 46R residues in PRRSV N protein were critical for induction of Tregs proliferation. The phenotype of induced Tregs closely resembled that of transforming-growth-factor-β-secreting T helper 3 Tregs in swine. These data should be useful for understanding the mechanism of immunity to PRRSV and development of infection control strategies in the future.
Collapse
Affiliation(s)
- Baochao Fan
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Xing Liu
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Juan Bai
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Yufeng Li
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Qiaoya Zhang
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Ping Jiang
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
- * E-mail:
| |
Collapse
|
50
|
Honeybee (Apis mellifera) Venom Reinforces Viral Clearance during the Early Stage of Infection with Porcine Reproductive and Respiratory Syndrome Virus through the Up-Regulation of Th1-Specific Immune Responses. Toxins (Basel) 2015; 7:1837-53. [PMID: 26008237 PMCID: PMC4448177 DOI: 10.3390/toxins7051837] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Revised: 05/15/2015] [Accepted: 05/18/2015] [Indexed: 02/06/2023] Open
Abstract
Porcine reproductive and respiratory syndrome (PRRS) is a chronic and immunosuppressive viral disease that is responsible for substantial economic losses for the swine industry. Honeybee venom (HBV) is known to possess several beneficial biological properties, particularly, immunomodulatory effects. Therefore, this study aimed at evaluating the effects of HBV on the immune response and viral clearance during the early stage of infection with porcine reproductive and respiratory syndrome virus (PRRSV) in pigs. HBV was administered via three routes of nasal, neck, and rectal and then the pigs were inoculated with PRRSV intranasally. The CD4+/CD8+ cell ratio and levels of interferon (IFN)-γ and interleukin (IL)-12 were significantly increased in the HBV-administered healthy pigs via nasal and rectal administration. In experimentally PRRSV-challenged pigs with virus, the viral genome load in the serum, lung, bronchial lymph nodes and tonsil was significantly decreased, as was the severity of interstitial pneumonia, in the nasal and rectal administration group. Furthermore, the levels of Th1 cytokines (IFN-γ and IL-12) were significantly increased, along with up-regulation of pro-inflammatory cytokines (TNF-α and IL-1β) with HBV administration. Thus, HBV administration—especially via the nasal or rectal route—could be a suitable strategy for immune enhancement and prevention of PRRSV infection in pigs.
Collapse
|