51
|
Dong VH, Tu PY, Tsai PC, Lin YH, Chang HL, Kuo TY, Chiou MT, Lin CN, Chung WB. Expression of Toll-like receptor signaling-related genes in pigs co-infected with porcine reproductive and respiratory syndrome virus and porcine circovirus type 2. Res Vet Sci 2015; 101:180-6. [PMID: 26022073 DOI: 10.1016/j.rvsc.2015.05.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Revised: 04/16/2015] [Accepted: 05/10/2015] [Indexed: 10/23/2022]
Abstract
Pigs co-infected with porcine reproductive and respiratory syndrome virus (PRRSV) and porcine circovirus type 2 (PCV2) have been shown to develop more severe diseases than pigs infected with PRRSV or PCV2 only. The underlying interaction mechanisms between the two viruses in developing the disease are unclear. The present study investigates the mRNA expression of Toll-like receptor (TLR) signaling-related molecules in peripheral blood mononuclear cells from pigs infected with PRRSV or PCV2 or both. The mRNA expression levels were determined by quantitative real-time RT-PCR. Co-infection of pigs with PRRSV and PCV2 resulted in a negatively synergistic effect on the mRNA expression of the negative regulators of TLR, including A20, Bcl-3, IRAK-M, MKP-1, SARM1 and SIGIRR, as well as the TLR downstream transcription factors IRF-1 and IRF-3. A positively synergistic effect of a combined infection of PRRSV and PCV2 on the CD14 mRNA expression was also observed.
Collapse
Affiliation(s)
- Van Hieu Dong
- Department of Veterinary Medicine, National Pingtung University of Science and Technology, Neipu, Pingtung 912, Taiwan, ROC
| | - Pang-Yan Tu
- Department of Veterinary Medicine, National Pingtung University of Science and Technology, Neipu, Pingtung 912, Taiwan, ROC
| | - Pei-Chun Tsai
- Department of Veterinary Medicine, National Pingtung University of Science and Technology, Neipu, Pingtung 912, Taiwan, ROC
| | - Yi-Hsin Lin
- Department of Veterinary Medicine, National Pingtung University of Science and Technology, Neipu, Pingtung 912, Taiwan, ROC
| | - Hsiu-Luan Chang
- Department of Animal Science, National Pingtung University of Science and Technology, Neipu, Pingtung 912, Taiwan, ROC
| | - Tsun-Yung Kuo
- Department of Biotechnology and Animal Science, National Ilan University, Ilan 26041, Taiwan, ROC
| | - Ming-Tang Chiou
- Department of Veterinary Medicine, National Pingtung University of Science and Technology, Neipu, Pingtung 912, Taiwan, ROC
| | - Chao-Nan Lin
- Department of Veterinary Medicine, National Pingtung University of Science and Technology, Neipu, Pingtung 912, Taiwan, ROC.
| | - Wen-Bin Chung
- Department of Veterinary Medicine, National Pingtung University of Science and Technology, Neipu, Pingtung 912, Taiwan, ROC.
| |
Collapse
|
52
|
Salguero FJ, Frossard JP, Rebel JMJ, Stadejek T, Morgan SB, Graham SP, Steinbach F. Host-pathogen interactions during porcine reproductive and respiratory syndrome virus 1 infection of piglets. Virus Res 2015; 202:135-43. [PMID: 25559070 PMCID: PMC7172408 DOI: 10.1016/j.virusres.2014.12.026] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Revised: 12/18/2014] [Accepted: 12/22/2014] [Indexed: 01/26/2023]
Abstract
Porcine reproductive and respiratory syndrome (PRRS) is a major disease affecting pigs worldwide and resulting in considerable economic losses. While PRRS is a global phenomenon, the causative viruses PRRSV-1 (first detected in Europe) and PRRSV-2 (isolated in North America) are genetically and biologically distinct. In addition, the disease outcome is directly linked to co-infections associated with the porcine respiratory disease complex and the host response is variable between different breeds of pigs. It is therefore warranted when studying the pathogenesis of PRRS to consider each viral genotype separately and apply careful consideration to the disease model studied. We here review the respiratory pig model for PRRSV-1, with a focus on a recent set of studies conducted with carefully selected virus strains and pigs, which may serve as both a baseline and benchmark for future investigation.
Collapse
Affiliation(s)
- Francisco J Salguero
- Virology Department, Animal and Plant Health Agency, Weybridge, Addlestone, United Kingdom; Department of Pathology and Infectious Diseases, School of Veterinary Medicine, University of Surrey, Guildford, United Kingdom
| | - Jean-Pierre Frossard
- Virology Department, Animal and Plant Health Agency, Weybridge, Addlestone, United Kingdom.
| | - Johanna M J Rebel
- Department of Infection Biology, Central Veterinary Institute, Lelystad, The Netherlands
| | - Tomasz Stadejek
- Department of Pathology and Veterinary Diagnostics, Faculty of Veterinary Medicine, Warsaw University of Life, Poland
| | - Sophie B Morgan
- Virology Department, Animal and Plant Health Agency, Weybridge, Addlestone, United Kingdom
| | - Simon P Graham
- Virology Department, Animal and Plant Health Agency, Weybridge, Addlestone, United Kingdom; Department of Pathology and Infectious Diseases, School of Veterinary Medicine, University of Surrey, Guildford, United Kingdom
| | - Falko Steinbach
- Virology Department, Animal and Plant Health Agency, Weybridge, Addlestone, United Kingdom; Department of Pathology and Infectious Diseases, School of Veterinary Medicine, University of Surrey, Guildford, United Kingdom
| |
Collapse
|
53
|
Zhang Q, Yoo D. PRRS virus receptors and their role for pathogenesis. Vet Microbiol 2015; 177:229-41. [PMID: 25912022 DOI: 10.1016/j.vetmic.2015.04.002] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Revised: 03/25/2015] [Accepted: 04/01/2015] [Indexed: 02/09/2023]
Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) is endemic in most pig producing countries worldwide and causes enormous economic losses to the swine industry. Specifically differentiated porcine alveolar macrophages are the primary target for PRRSV infection in pigs. At least six cellular molecules have been described so far as putative receptors for PRRSV, and they include heparan sulfate, vimentin, CD151, sialoadhesin (CD169; siglec-1), dendritic cell-specific intercellular adhesion melecule-3-grabbing non-integrin (DC-SIGN; CD209), and CD163 (SRCR, cysteine-rich scavenger receptor). Progress has been made to shed light on the interactions between cells and PRRSV, and this review describes the advances and current understanding of the entry of PRRSV to cells with a particular focus on the role of CD163 and sialoadhesin for infection and PRRSV pathogenesis. CD163 is most likely the primary and core receptor for PRRSV and determines the susceptibility of cells to the virus. Sialoadhesin is either unnecessary for infection or may function as an accessory protein. Sialoadhesin has been mostly studied for genotype I PRRSV whereas the utilization of CD163 has been mostly studied using genotype II PRRSV, and whether each genotype indeed utilizes a different receptor is unclear.
Collapse
Affiliation(s)
- Qingzhan Zhang
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Dongwan Yoo
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, IL, United States.
| |
Collapse
|
54
|
García-Nicolás O, Rosales RS, Pallarés FJ, Risco D, Quereda JJ, Graham SP, Frossard JP, Morgan SB, Steinbach F, Drew TW, Strickland TS, Salguero FJ. Comparative analysis of cytokine transcript profiles within mediastinal lymph node compartments of pigs after infection with porcine reproductive and respiratory syndrome genotype 1 strains differing in pathogenicity. Vet Res 2015; 46:34. [PMID: 25889072 PMCID: PMC4364558 DOI: 10.1186/s13567-015-0161-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Accepted: 02/06/2015] [Indexed: 01/28/2023] Open
Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) induces a weak immune response enabling it to persist in different organs of infected pigs. This has been attributed to the ability of PRRSV to influence the induction of cytokine responses. In this study, we investigated the cytokine transcriptional profiles in different compartments of the mediastinal lymph node of pigs infected with three genotype 1 PRRSV strains of differing pathogenicity: the low virulence prototype Lelystad virus (LV), and UK field strain 215–06 and the highly virulent subtype 3 SU1-Bel isolate from Belarus. We have used a combination of laser capture micro-dissection (LCM) followed by real time quantitative PCR (RT-qPCR) and immunohistochemical (IHC) detection of immune cell markers (CD3, CD79a and MAC387) and RT-qPCR quantification of PRRSV and cytokine transcripts. Compared to mock infected pigs, we found a significant downregulation of TNF-α and IFN-α in follicular and interfollicular areas of the mediastinal lymph node from 3 days post-infection (dpi) in animals infected with all three strains. This was accompanied by a transient B cell depletion and T cell and macrophage infiltration in the follicles together with T cell depletion in the interfollicular areas. A delayed upregulation of IFN-γ and IL-23p19 was observed mainly in the follicles. The PRRSV load was higher in all areas and time-points studied in the animals infected with the SU1-Bel strain. This paper describes the first application of LCM to study the cytokine transcript profiles and virus distribution in different compartments of the lymph node of pigs.
Collapse
Affiliation(s)
- Obdulio García-Nicolás
- Department of Anatomy and Comparative Pathology, Faculty of Veterinary Medicine, Murcia University, "Mare Nostrum Excellence Campus - 3738", 30100, Murcia, Spain.
| | - Rubén S Rosales
- Department of Bacteriology, Animal and Plant Health Agency, Addlestone, Surrey, KT15 3NB, UK.
| | - Francisco J Pallarés
- Department of Anatomy and Comparative Pathology, Faculty of Veterinary Medicine, Murcia University, "Mare Nostrum Excellence Campus - 3738", 30100, Murcia, Spain.
| | - David Risco
- Red de Grupos de Investigación Recursos Faunísticos, Facultad de Veterinaria, Universidad de Extremadura, 10003, Cáceres, Spain.
| | - Juan J Quereda
- Spanish National Center of Biotechnology (CSIC), C/Darwin 3, Campus de Cantoblanco, 28049, Madrid, Spain.
| | - Simon P Graham
- Department of Virology, Animal and Plant Health Agency, Addlestone, Surrey, KT15 3NB, UK.
| | - Jean-Pierre Frossard
- Department of Virology, Animal and Plant Health Agency, Addlestone, Surrey, KT15 3NB, UK.
| | - Sophie B Morgan
- Department of Virology, Animal and Plant Health Agency, Addlestone, Surrey, KT15 3NB, UK.
| | - Falko Steinbach
- Department of Virology, Animal and Plant Health Agency, Addlestone, Surrey, KT15 3NB, UK. .,Department of Pathology and Infectious Diseases, School of Veterinary Medicine, University of Surrey, Guildford, GU2 7TE, UK.
| | - Trevor W Drew
- Department of Virology, Animal and Plant Health Agency, Addlestone, Surrey, KT15 3NB, UK.
| | - Tony S Strickland
- Department of Pathology, Animal and Plant Health Agency, Addlestone, KT15 3NB, UK.
| | - Francisco J Salguero
- Department of Pathology, Animal and Plant Health Agency, Addlestone, KT15 3NB, UK. .,Department of Pathology and Infectious Diseases, School of Veterinary Medicine, University of Surrey, Guildford, GU2 7TE, UK.
| |
Collapse
|
55
|
He Q, Li Y, Zhou L, Ge X, Guo X, Yang H. Both Nsp1β and Nsp11 are responsible for differential TNF-α production induced by porcine reproductive and respiratory syndrome virus strains with different pathogenicity in vitro. Virus Res 2015; 201:32-40. [PMID: 25708177 DOI: 10.1016/j.virusres.2015.02.014] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Revised: 02/11/2015] [Accepted: 02/13/2015] [Indexed: 11/18/2022]
Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) has been recognized to be one of the most important pathogens severely affecting global swine industry. An increasingly number of studies have paid much attention to the diverse roles of its nonstructural proteins (Nsps) in regulating the innate immune response of host upon PRRSV infection. In the present study, we first discovered that highly pathogenic PRRSV (HP-PRRSV) and low pathogenic PRRSV (LP-PRRSV) infection exhibited a differential TNF-α expression in pulmonary alveolar macrophages (PAMs), showing that HP-PRRSV infection induces lower TNF-α production at protein level in PAMs, compared with LP-PRRSV. Next, HP-PRRSV was confirmed to strongly suppress TNF-α production by inhibiting ERK signaling pathway. Finally, both Nsp1β and Nsp11 were demonstrated to be responsible for the inhibitory effect on TNF-α production induced by HP-PRRSV and the differential TNF-α production in PAMs. These findings contribute to the understanding of the pathogenesis of the Chinese HP-PRRSV.
Collapse
Affiliation(s)
- Qing He
- Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine and State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing 100193, People's Republic of China
| | - Yan Li
- Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine and State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing 100193, People's Republic of China
| | - Lei Zhou
- Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine and State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing 100193, People's Republic of China
| | - Xinna Ge
- Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine and State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing 100193, People's Republic of China
| | - Xin Guo
- Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine and State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing 100193, People's Republic of China.
| | - Hanchun Yang
- Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine and State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing 100193, People's Republic of China.
| |
Collapse
|
56
|
Amarilla SP, Gómez-Laguna J, Carrasco L, Rodríguez-Gómez IM, Caridad Y Ocerín JM, Morgan SB, Graham SP, Frossard JP, Drew TW, Salguero FJ. A comparative study of the local cytokine response in the lungs of pigs experimentally infected with different PRRSV-1 strains: upregulation of IL-1α in highly pathogenic strain induced lesions. Vet Immunol Immunopathol 2015; 164:137-47. [PMID: 25739319 DOI: 10.1016/j.vetimm.2015.02.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Revised: 01/06/2015] [Accepted: 02/05/2015] [Indexed: 11/17/2022]
Abstract
Porcine reproductive and respiratory syndrome viruses (PRRSV) show high genetic differences both among and within genotypes. Recently, several highly pathogenic PRRSV (HP-PRRSV) strains have been described. This study compares and characterizes the production of cytokines by pulmonary macrophages in pigs experimentally infected with four different PRRSV-1 strains: two low-virulent strains, Lelystad (LV) and a British field strain (215-06); a HP strain (SU1-bel) from Belarus and the attenuated vaccine strain DV (Porcilis(®) PRRS). Animals were clinically monitored and post-mortem examinations were performed at 3, 7 and 35 days post-infection (dpi). Lung samples were processed for histopathological and immunohistochemical studies by using specific antibodies against PRRSV, IL1-α, IL-6, TNF-α, IL-10 and IFN-γ. SU1-bel infected animals presented the highest mean scores for clinical observations, gross and microscopic lesions as well as for PRRSV expression compared with the other infected groups (p≤0.027). These animals displayed the highest expression of IL1-α at 7dpi, together with the highest score for lung pathology, whereas LV, 215-06 and DV inoculated animals only showed a transient enhancement in some of these cytokines. SU1-bel-infected pigs showed a positive correlation between the amount of PRRSV antigen and IL-1α expression (r=0.645, p<0.001). The highest expression of IL-10 was detected in 215-06-infected animals (p≤0.004), with a positive correlation with the numbers of virus-infected cells (r=0.375, p≤0.013). In conclusion, the HP-PRRSV SU1-bel strain replicated more efficiently in the lung of infected animals and induced a higher expression of IL-1α than the other PRRSV-1-infected groups, which may have played a key role in the onset of the clinical signs and interstitial pneumonia.
Collapse
Affiliation(s)
- Shyrley P Amarilla
- Department of Anatomy and Comparative Pathology, Faculty of Veterinary Medicine, Campus Universitario de Rabanales, 'International Excellence Agrifood Campus, CeiA3', 14071 Cordoba, Spain.
| | | | - Librado Carrasco
- Department of Anatomy and Comparative Pathology, Faculty of Veterinary Medicine, Campus Universitario de Rabanales, 'International Excellence Agrifood Campus, CeiA3', 14071 Cordoba, Spain
| | - Irene M Rodríguez-Gómez
- Department of Anatomy and Comparative Pathology, Faculty of Veterinary Medicine, Campus Universitario de Rabanales, 'International Excellence Agrifood Campus, CeiA3', 14071 Cordoba, Spain
| | - José M Caridad Y Ocerín
- Department of Statistics, Econometrics, Operations Research, Business Organization and Applied Economics, Faculty of Law and Economics, 14071 Cordoba, Spain
| | - Sophie B Morgan
- Department of Virology, Animal Health and Veterinary Laboratories Agency-Weybridge, Addlestone, Surrey KT15 3NB, UK
| | - Simon P Graham
- Department of Virology, Animal Health and Veterinary Laboratories Agency-Weybridge, Addlestone, Surrey KT15 3NB, UK
| | - Jean-Pierre Frossard
- Department of Virology, Animal Health and Veterinary Laboratories Agency-Weybridge, Addlestone, Surrey KT15 3NB, UK
| | - Trevor W Drew
- Department of Virology, Animal Health and Veterinary Laboratories Agency-Weybridge, Addlestone, Surrey KT15 3NB, UK
| | - Francisco J Salguero
- Department of Virology, Animal Health and Veterinary Laboratories Agency-Weybridge, Addlestone, Surrey KT15 3NB, UK; Department of Pathology and Infectious Diseases, School of Veterinary Medicine, University of Surrey, Guildford GU2 7TE, UK
| |
Collapse
|
57
|
Marco-Ramell A, Hummel K, Razzazi-Fazeli E, Bassols A, Miller I. Concentration and pattern changes of porcine serum apolipoprotein A-I in four different infectious diseases. Electrophoresis 2015; 36:543-51. [PMID: 25377528 DOI: 10.1002/elps.201400299] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Revised: 10/03/2014] [Accepted: 10/29/2014] [Indexed: 01/04/2023]
Abstract
Apolipoprotein A-I (Apo A-I) is a major protein in lipid/lipoprotein metabolism and decreased serum levels have been observed in many species in response to inflammatory and infectious challenges. Little is known about the porcine homologue, therefore in this work we have characterized it through biochemical and proteomic techniques. In 2DE, porcine serum Apo A-I is found as three spots, the two more acidic ones corresponding to the mature protein, the more basic spot to the protein precursor. Despite high sequence coverage in LC-MS/MS, we did not find a sequence or PTM difference between the two mature protein species. Besides this biochemical characterization, we measured overall levels and relative species abundance of serum Apo A-I in four different viral and bacterial porcine infectious diseases. Lower overall amounts of Apo A-I were observed in Salmonella typhimurium and Escherichia coli infections. In the 2DE protein pattern, an increase of the protein precursor together with a lower level of mature protein species were detected in the porcine circovirus type 2-systemic disease and S. typhimurium infection. These results reveal that both the porcine serum Apo A-I concentration and the species pattern are influenced by the nature of the infectious disease.
Collapse
Affiliation(s)
- Anna Marco-Ramell
- Departament de Bioquímica i Biologia Molecular, Facultat de Veterinària, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | | | | | | | | |
Collapse
|
58
|
Pileri E, Gibert E, Soldevila F, García-Saenz A, Pujols J, Diaz I, Darwich L, Casal J, Martín M, Mateu E. Vaccination with a genotype 1 modified live vaccine against porcine reproductive and respiratory syndrome virus significantly reduces viremia, viral shedding and transmission of the virus in a quasi-natural experimental model. Vet Microbiol 2014; 175:7-16. [PMID: 25439650 DOI: 10.1016/j.vetmic.2014.11.007] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Revised: 11/03/2014] [Accepted: 11/06/2014] [Indexed: 11/16/2022]
Abstract
The present study assessed the efficacy of vaccination against genotype 1 porcine reproductive and respiratory syndrome virus (PRRSV) in terms of reduction of the transmission. Ninety-eight 3-week-old piglets were divided in two groups: V (n=40) and NV (n=58) that were housed separately. V animals were vaccinated with a commercial genotype 1 PRRSV vaccine while NV were kept as controls. On day 35 post-vaccination, 14 NV pigs were separated and inoculated intranasally with 2 ml of a heterologous genotype 1 PRRSV isolate ("seeder" pigs, SP). The other V and NV animals were distributed in groups of 5 pigs each. Two days later, one SP was introduced into each pen to expose V and NV to PRRSV. Sentinel pigs were allocated in adjacent pens. Follow-up was of 21 days. All NV (30/30) became viremic after contact with SP while only 53% of V pigs were detected so (21/40, p<0.05). Vaccination shortened viremia (12.2±4 versus 3.7±3.4 days in NV and V pigs, respectively, p<0.01). The 50% survival time for becoming infected (Kaplan-Meier) for V was 21 days (CI95%=14.1-27.9) compared to 7 days (CI95%=5.2-8.7) for NV animals (p<0.01). These differences were reflected in the R value as well: 2.78 (CI95%=2.13-3.43) for NV and 0.53 (CI95%=0.19-0.76) for V pigs (p<0.05). All sentinel pigs (10/10) in pens adjacent to NV+SP pens got infected compared to 1/4 sentinel pigs allocated contiguous to a V+SP pen. These data show that vaccination of piglets significantly decrease parameters related to PRRSV transmission.
Collapse
Affiliation(s)
- Emanuela Pileri
- Departament de Sanitat i Anatomia Animals, Universitat Autònoma de Barcelona, 08193 Bellaterra, Cerdanyola del Vallès, Spain; Center de Recerca en Sanitat Animal (CReSA), UAB-IRTA, Campus de la Universitat Autònoma de Barcelona, 08193 Bellaterra, Cerdanyola del Vallès, Spain.
| | - Elisa Gibert
- Center de Recerca en Sanitat Animal (CReSA), UAB-IRTA, Campus de la Universitat Autònoma de Barcelona, 08193 Bellaterra, Cerdanyola del Vallès, Spain.
| | - Ferran Soldevila
- Center de Recerca en Sanitat Animal (CReSA), UAB-IRTA, Campus de la Universitat Autònoma de Barcelona, 08193 Bellaterra, Cerdanyola del Vallès, Spain.
| | - Ariadna García-Saenz
- Center de Recerca en Sanitat Animal (CReSA), UAB-IRTA, Campus de la Universitat Autònoma de Barcelona, 08193 Bellaterra, Cerdanyola del Vallès, Spain.
| | - Joan Pujols
- Center de Recerca en Sanitat Animal (CReSA), UAB-IRTA, Campus de la Universitat Autònoma de Barcelona, 08193 Bellaterra, Cerdanyola del Vallès, Spain.
| | - Ivan Diaz
- Center de Recerca en Sanitat Animal (CReSA), UAB-IRTA, Campus de la Universitat Autònoma de Barcelona, 08193 Bellaterra, Cerdanyola del Vallès, Spain.
| | - Laila Darwich
- Departament de Sanitat i Anatomia Animals, Universitat Autònoma de Barcelona, 08193 Bellaterra, Cerdanyola del Vallès, Spain; Center de Recerca en Sanitat Animal (CReSA), UAB-IRTA, Campus de la Universitat Autònoma de Barcelona, 08193 Bellaterra, Cerdanyola del Vallès, Spain.
| | - Jordi Casal
- Departament de Sanitat i Anatomia Animals, Universitat Autònoma de Barcelona, 08193 Bellaterra, Cerdanyola del Vallès, Spain; Center de Recerca en Sanitat Animal (CReSA), UAB-IRTA, Campus de la Universitat Autònoma de Barcelona, 08193 Bellaterra, Cerdanyola del Vallès, Spain.
| | - Marga Martín
- Departament de Sanitat i Anatomia Animals, Universitat Autònoma de Barcelona, 08193 Bellaterra, Cerdanyola del Vallès, Spain; Center de Recerca en Sanitat Animal (CReSA), UAB-IRTA, Campus de la Universitat Autònoma de Barcelona, 08193 Bellaterra, Cerdanyola del Vallès, Spain.
| | - Enric Mateu
- Departament de Sanitat i Anatomia Animals, Universitat Autònoma de Barcelona, 08193 Bellaterra, Cerdanyola del Vallès, Spain; Center de Recerca en Sanitat Animal (CReSA), UAB-IRTA, Campus de la Universitat Autònoma de Barcelona, 08193 Bellaterra, Cerdanyola del Vallès, Spain.
| |
Collapse
|
59
|
Amadori M, Razzuoli E. Immune Control of PRRS: Lessons to be Learned and Possible Ways Forward. Front Vet Sci 2014; 1:2. [PMID: 26664910 PMCID: PMC4668844 DOI: 10.3389/fvets.2014.00002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Accepted: 05/19/2014] [Indexed: 12/29/2022] Open
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. The disease can be controlled by farm management programs, which can be supported by vaccination or conditioning of animals to circulating PRRS virus (PRRSV) strains. Yet, PRRS still represents a cause of heavy losses for the pig industry worldwide. Immunological control strategies are often compounded by poor and late development of adaptive immunity in both vaccinated and infected animals. Also, there is evidence that results of field trials can be worse than those of experimental studies in isolation facilities. Neutralizing antibody (NA) was shown to prevent PRRSV infection. Instead, the role of NA and adaptive immunity on the whole in virus clearance after established PRRSV infections is still contentious. Pigs eventually eliminate PRRSV infection, which may be correlated with an “educated,” innate immune response, which may also develop following vaccination. In addition to vaccination, an immunomodulation strategy for PRRS can be reasonably advocated in pig “problem” farms, where a substantial control of disease prevalence and disease-related losses is badly needed. This is not at odds with vaccination, which should be preferably restricted to PRRSV-free animals bound for PRRSV-infected farm units. Oral, low-dose, interferon-α treatments proved effective on farm for the control of respiratory and reproductive disease outbreaks, whereas the results were less clear in isolation facilities. Having in mind the crucial interaction between PRRSV and bacterial lipopolysaccharides for occurrence of respiratory disease, the strong control actions of low-dose type I interferons on the inflammatory response observed in vitro and in vivo probably underlie the rapid clinical responses observed in field trials.
Collapse
Affiliation(s)
- Massimo Amadori
- Laboratory of Cellular Immunology, Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia Romagna , Brescia , Italy
| | - Elisabetta Razzuoli
- Laboratory of Cellular Immunology, Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia Romagna , Brescia , Italy
| |
Collapse
|
60
|
Han M, Yoo D. Modulation of innate immune signaling by nonstructural protein 1 (nsp1) in the family Arteriviridae. Virus Res 2014; 194:100-9. [PMID: 25262851 PMCID: PMC7114407 DOI: 10.1016/j.virusres.2014.09.007] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Revised: 09/16/2014] [Accepted: 09/17/2014] [Indexed: 12/24/2022]
Abstract
Arteriviruses infect immune cells and may cause persistence in infected hosts. Inefficient induction of pro-inflammatory cytokines and type I IFNs are observed during infection of this group of viruses, suggesting that they may have evolved to escape the host immune surveillance for efficient survival. Recent studies have identified viral proteins regulating the innate immune signaling, and among these, nsp1 (nonstructural protein 1) is the most potent IFN antagonist. For porcine reproductive and respiratory syndrome virus (PRRSV), individual subunits (nsp1α and nsp1β) of nsp1 suppress type I IFN production. In particular, PRRSV-nsp1α degrades CREB (cyclic AMP responsive element binding)-binding protein (CBP), a key component of the IFN enhanceosome, whereas PRRSV-nsp1β degrades karyopherin-α1 which is known to mediate the nuclear import of ISGF3 (interferon-stimulated gene factor 3). All individual subunits of nsp1 of PRRSV, equine arteritis virus (EAV), lactate dehydrogenase-elevating virus (LDV), and simian hemorrhagic fever virus (SHFV) appear to contain IFN suppressive activities. As with PRRSV-nsp1α, CBP degradation is evident by LDV-nsp1α and partly by SHFV-nsp1γ. This review summarizes the biogenesis and the role of individual subunits of nsp1 of arteriviruses for innate immune modulation.
Collapse
Affiliation(s)
- Mingyuan Han
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, IL 61802, USA
| | - Dongwan Yoo
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, IL 61802, USA.
| |
Collapse
|
61
|
Go N, Bidot C, Belloc C, Touzeau S. Integrative model of the immune response to a pulmonary macrophage infection: what determines the infection duration? PLoS One 2014; 9:e107818. [PMID: 25233096 PMCID: PMC4169448 DOI: 10.1371/journal.pone.0107818] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2014] [Accepted: 08/09/2014] [Indexed: 12/23/2022] Open
Abstract
The immune mechanisms which determine the infection duration induced by pathogens targeting pulmonary macrophages are poorly known. To explore the impact of such pathogens, it is indispensable to integrate the various immune mechanisms and to take into account the variability in pathogen virulence and host susceptibility. In this context, mathematical models complement experimentation and are powerful tools to represent and explore the complex mechanisms involved in the infection and immune dynamics. We developed an original mathematical model in which we detailed the interactions between the macrophages and the pathogen, the orientation of the adaptive response and the cytokine regulations. We applied our model to the Porcine Respiratory and Reproductive Syndrome virus (PRRSv), a major concern for the swine industry. We extracted value ranges for the model parameters from modelling and experimental studies on respiratory pathogens. We identified the most influential parameters through a sensitivity analysis. We defined a parameter set, the reference scenario, resulting in a realistic and representative immune response to PRRSv infection. We then defined scenarios corresponding to graduated levels of strain virulence and host susceptibility around the reference scenario. We observed that high levels of antiviral cytokines and a dominant cellular response were associated with either short, the usual assumption, or long infection durations, depending on the immune mechanisms involved. To identify these mechanisms, we need to combine the levels of antiviral cytokines, including , and . The latter is a good indicator of the infected macrophage level, both combined provide the adaptive response orientation. Available PRRSv vaccines lack efficiency. By integrating the main interactions between the complex immune mechanisms, this modelling framework could be used to help designing more efficient vaccination strategies.
Collapse
Affiliation(s)
- Natacha Go
- UR341 MIA, INRA, Jouy-en-Josas, France
- LUNAM Université, Oniris, INRA UMR 1300 BioEpAR, Nantes, France
- * E-mail:
| | | | | | - Suzanne Touzeau
- UMR1355 ISA, INRA, Université Nice Sophia Antipolis, CNRS, Sophia Antipolis, France
- BIOCORE, Inria, Sophia Antipolis, France
| |
Collapse
|
62
|
Li Y, Zhou L, Zhang J, Ge X, Zhou R, Zheng H, Geng G, Guo X, Yang H. Nsp9 and Nsp10 contribute to the fatal virulence of highly pathogenic porcine reproductive and respiratory syndrome virus emerging in China. PLoS Pathog 2014; 10:e1004216. [PMID: 24992286 PMCID: PMC4081738 DOI: 10.1371/journal.ppat.1004216] [Citation(s) in RCA: 137] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2014] [Accepted: 05/15/2014] [Indexed: 11/19/2022] Open
Abstract
Atypical porcine reproductive and respiratory syndrome (PRRS), which is caused by the Chinese highly pathogenic PRRS virus (HP-PRRSV), has resulted in large economic loss to the swine industry since its outbreak in 2006. However, to date, the region(s) within the viral genome that are related to the fatal virulence of HP-PRRSV remain unknown. In the present study, we generated a series of full-length infectious cDNA clones with swapped coding regions between the highly pathogenic RvJXwn and low pathogenic RvHB-1/3.9. Next, the in vitro and in vivo replication and pathogenicity for piglets of the rescued chimeric viruses were systematically analyzed and compared with their backbone viruses. First, we swapped the regions including the 5′UTR+ORF1a, ORF1b, and structural proteins (SPs)-coding region between the two viruses and demonstrated that the nonstructural protein-coding region, ORF1b, is directly related to the fatal virulence and increased replication efficiency of HP-PRRSV both in vitro and in vivo. Furthermore, we substituted the nonstructural protein (Nsp) 9-, Nsp10-, Nsp11- and Nsp12-coding regions separately; or Nsp9- and Nsp10-coding regions together; or Nsp9-, Nsp10- and Nsp11-coding regions simultaneously between the two viruses. Our results indicated that the HP-PRRSV Nsp9- and Nsp10-coding regions together are closely related to the replication efficiency in vitro and in vivo and are related to the increased pathogenicity and fatal virulence for piglets. Our findings suggest that Nsp9 and Nsp10 together contribute to the fatal virulence of HP-PRRSV emerging in China, helping to elucidate the pathogenesis of this virus. PRRS is a considerable threat to the pig industry worldwide. A large-scale atypical PRRS caused by highly pathogenic PRRSV (HP-PRRSV) that emerged in 2006 has resulted in considerable economic loss to Chinese pig production. The disease is characterized by a high body temperature (41°C–42°C), morbidity and by mortality of the affected pigs. Although the genomic marker, the 30-amino-acid deletion in its Nsp2-coding region has been previously verified to have no relation to its increased pathogenicity, the genomic region(s) associated with the fatal virulence of HP-PRRSV remain unclear. A series of chimeric viruses with swapped coding regions between HP- and LP-PRRSV were constructed, and their growth abilities and pathogenicities in piglets were analyzed. Our results demonstrated that Nsp9 and Nsp10 together contribute to the replication efficiency and the fatal virulence of HP-PRRSV for piglets. Our finding is not only the first unambiguous illumination concerning the key virulence determinant of Chinese HP-PRRSV but it also provides a novel insight for understanding the molecular pathogenesis of this virus and for designing new drugs and vaccines against PRRSV infection in the future.
Collapse
Affiliation(s)
- Yan Li
- Key Laboratory of Animal Epidemiology and Zoonosis of the Ministry of Agriculture, College of Veterinary Medicine and State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, People's Republic of China
| | - Lei Zhou
- Key Laboratory of Animal Epidemiology and Zoonosis of the Ministry of Agriculture, College of Veterinary Medicine and State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, People's Republic of China
| | - Jialong Zhang
- Key Laboratory of Animal Epidemiology and Zoonosis of the Ministry of Agriculture, College of Veterinary Medicine and State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, People's Republic of China
| | - Xinna Ge
- Key Laboratory of Animal Epidemiology and Zoonosis of the Ministry of Agriculture, College of Veterinary Medicine and State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, People's Republic of China
| | - Rong Zhou
- Key Laboratory of Animal Epidemiology and Zoonosis of the Ministry of Agriculture, College of Veterinary Medicine and State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, People's Republic of China
| | - Huaguo Zheng
- Key Laboratory of Animal Epidemiology and Zoonosis of the Ministry of Agriculture, College of Veterinary Medicine and State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, People's Republic of China
| | - Gang Geng
- Key Laboratory of Animal Epidemiology and Zoonosis of the Ministry of Agriculture, College of Veterinary Medicine and State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, People's Republic of China
| | - Xin Guo
- Key Laboratory of Animal Epidemiology and Zoonosis of the Ministry of Agriculture, College of Veterinary Medicine and State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, People's Republic of China
| | - Hanchun Yang
- Key Laboratory of Animal Epidemiology and Zoonosis of the Ministry of Agriculture, College of Veterinary Medicine and State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, People's Republic of China
- * E-mail:
| |
Collapse
|
63
|
García-Nicolás O, Quereda JJ, Gómez-Laguna J, Salguero FJ, Carrasco L, Ramis G, Pallarés FJ. Cytokines transcript levels in lung and lymphoid organs during genotype 1 Porcine Reproductive and Respiratory Syndrome Virus (PRRSV) infection. Vet Immunol Immunopathol 2014; 160:26-40. [DOI: 10.1016/j.vetimm.2014.03.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Revised: 03/10/2014] [Accepted: 03/18/2014] [Indexed: 12/24/2022]
|
64
|
Ren JQ, Sun WC, Lu HJ, Wen SB, Jing J, Yan FL, Liu H, Liu CX, Xiao PP, Chen X, Du SW, Du R, Jin NY. Construction and immunogenicity of a DNA vaccine coexpressing GP3 and GP5 of genotype-I porcine reproductive and respiratory syndrome virus. BMC Vet Res 2014; 10:128. [PMID: 24916952 PMCID: PMC4090398 DOI: 10.1186/1746-6148-10-128] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2013] [Accepted: 05/28/2014] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND The European (EU) genotype of porcine reproductive and respiratory syndrome virus (Genotype-I PRRSV) has recently emerged in China. The coexistence of Genotype-I and -II PRRSV strains could cause seriously affect PRRSV diagnosis and management. Current vaccines are not able to protect against PRRSV infection completely and have inherent drawbacks. Thus, genetically engineered vaccines, including DNA vaccine and live vector engineered vaccines, have been developed. This study aimed to determine the enhanced immune responses of mice inoculated with a DNA vaccine coexpressing GP3 and GP5 of a Genotype-I PRRSV. RESULTS To evaluate the immunogenicity of GP3 and GP5 proteins from European-type PRRSV, three DNA vaccines, pVAX1-EU-ORF3-ORF5, pVAX1-EU-ORF3 and pVAX1-EU-ORF5, were constructed, which were based on a Genotype-I LV strain (GenBank ID: M96262). BALB/c mice were immunized with the DNA vaccines; delivered in the form of chitosan-DNA nanoparticles. To increase the efficiency of the vaccine, Quil A (Quillaja) was used as an adjuvant. GP3 and GP5-specific antibodies, neutralizing antibodies and cytokines (IL-2, IL-4, IL-10 and IFN gamma) from the immunized mice sera, and other immune parameters, were examined, including T-cell proliferation responses and subgroups of spleen T-lymphocytes. The results showed that ORF3 and ORF5 proteins of Genotype-I PRRSV induced GP3 and GP5-specific antibodies that could neutralize the virus. The levels of Cytokines IL-2, IL-4, IL-10, and IFN-γ of the experimental groups were significantly higher than those of control groups after booster vaccination (P < 0.05). The production of CD3+CD4+ and CD3+CD8+ T lymphocyte was also induced. T lymphocyte proliferation assays showed that the PRRSV LV strain virus could stimulate the proliferation of T lymphocytes in mice in the experimental group. CONCLUSIONS Using Quil A as adjuvant, Genotype-I PRRSV GP3 and GP5 proteins produced good immunogenicity and reactivity. More importantly, better PRRSV-specific neutralizing antibody titers and cell-mediated immune responses were observed in mice immunized with the DNA vaccine co-expressing GP3 and GP5 proteins than in mice immunized with a DNA vaccine expressing either protein singly. The results of this study demonstrated that co-immunization with GP3 and GP5 produced a better immune response in mice.
Collapse
Affiliation(s)
- Jing-Qiang Ren
- Institute of Military Veterinary, Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Academy of Military Medical Sciences, Changchun 130122, China
- College of Veterinary Medicine, Jilin University, Changchun 130062, China
- Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Changchun 130122, China
| | - Wen-Chao Sun
- Institute of Military Veterinary, Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Academy of Military Medical Sciences, Changchun 130122, China
- College of Animal Science and Technology, Graduate School, Jilin Agricultural University, Changchun 130118, China
| | - Hui-Jun Lu
- Institute of Military Veterinary, Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Academy of Military Medical Sciences, Changchun 130122, China
| | - Shu-Bo Wen
- Institute of Military Veterinary, Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Academy of Military Medical Sciences, Changchun 130122, China
- College of Animal Science and Technology, Graduate School, Jilin Agricultural University, Changchun 130118, China
| | - Jie Jing
- Institute of Military Veterinary, Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Academy of Military Medical Sciences, Changchun 130122, China
- College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Fu-Long Yan
- Institute of Military Veterinary, Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Academy of Military Medical Sciences, Changchun 130122, China
- College of Animal Science and Technology, Graduate School, Jilin Agricultural University, Changchun 130118, China
| | - Hao Liu
- Institute of Military Veterinary, Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Academy of Military Medical Sciences, Changchun 130122, China
- Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Changchun 130122, China
| | - Cun-Xia Liu
- Institute of Military Veterinary, Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Academy of Military Medical Sciences, Changchun 130122, China
| | - Peng-Peng Xiao
- Institute of Military Veterinary, Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Academy of Military Medical Sciences, Changchun 130122, China
- College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Xing Chen
- Institute of Military Veterinary, Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Academy of Military Medical Sciences, Changchun 130122, China
- College of Animal Science and Technology, Graduate School, Jilin Agricultural University, Changchun 130118, China
| | - Shou-Wen Du
- Institute of Military Veterinary, Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Academy of Military Medical Sciences, Changchun 130122, China
- College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Rui Du
- College of Animal Science and Technology, Graduate School, Jilin Agricultural University, Changchun 130118, China
| | - Ning-Yi Jin
- Institute of Military Veterinary, Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Academy of Military Medical Sciences, Changchun 130122, China
- College of Veterinary Medicine, Jilin University, Changchun 130062, China
| |
Collapse
|
65
|
Rodríguez-Gómez IM, Barranco I, Amarilla SP, García-Nicolás O, Salguero FJ, Carrasco L, Gómez-Laguna J. Activation of extrinsic- and Daxx-mediated pathways in lymphoid tissue of PRRSV-infected pigs. Vet Microbiol 2014; 172:186-94. [PMID: 24939593 DOI: 10.1016/j.vetmic.2014.05.025] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Revised: 03/11/2014] [Accepted: 05/27/2014] [Indexed: 10/25/2022]
Abstract
Porcine Reproductive and Respiratory Syndrome Virus (PRRSV) is a major infectious pathogen in pigs leading to huge economical losses worldwide. PRRSV is able to escape from host immunity and causes transient infections. In the present study, expression of different apoptotic markers and its connection with PRRSV were assessed in tonsil and mediastinal lymph node from PRRSV-infected pigs. Cleaved caspase (CCasp)8, CCasp9, Fas, Daxx, CCasp3 and PRRSV expression were analyzed by immunohistochemistry. An up-regulation of CCasp8, Fas and CCasp3 expression in lymphocytes and macrophages from both organs was found during PRRSV infection, indicating the activation of the extrinsic-mediated pathway of apoptosis. Moreover, Daxx expression was also enhanced in macrophages of both organs, suggesting a simultaneous caspase-independent pathway of apoptosis. A correlation between the expression of the different apoptotic markers and IL-10, IL-6 and TGF-β but not with PRRSV antigen was found in our study, which supports the hypothesis of an indirect mechanism in PRRSV-induced apoptosis.
Collapse
Affiliation(s)
- Irene M Rodríguez-Gómez
- Department of Anatomy and Comparative Pathology, Faculty of Veterinary Medicine, Córdoba University, "International Excellence Agrifood Campus - CeiA3", 14014 Córdoba, Spain.
| | - Inmaculada Barranco
- Department of Anatomy and Comparative Pathology, Faculty of Veterinary Medicine, Córdoba University, "International Excellence Agrifood Campus - CeiA3", 14014 Córdoba, Spain
| | - Shyrley P Amarilla
- Department of Anatomy and Comparative Pathology, Faculty of Veterinary Medicine, Córdoba University, "International Excellence Agrifood Campus - CeiA3", 14014 Córdoba, Spain
| | - Obdulio García-Nicolás
- Department of Anatomy and Comparative Pathology, Faculty of Veterinary Medicine, Murcia University, "Mare Nostrum Campus of Excellence - 37 38", 30100 Murcia, Spain
| | - Francisco J Salguero
- School of Veterinary Medicine, Faculty of Health & Medical Sciences, Surrey University, Guildford, Surrey, GU2 7TE, United Kingdom
| | - Librado Carrasco
- Department of Anatomy and Comparative Pathology, Faculty of Veterinary Medicine, Córdoba University, "International Excellence Agrifood Campus - CeiA3", 14014 Córdoba, Spain
| | | |
Collapse
|
66
|
Kuzemtseva L, de la Torre E, Martín G, Soldevila F, Ait-Ali T, Mateu E, Darwich L. Regulation of toll-like receptors 3, 7 and 9 in porcine alveolar macrophages by different genotype 1 strains of porcine reproductive and respiratory syndrome virus. Vet Immunol Immunopathol 2014; 158:189-98. [DOI: 10.1016/j.vetimm.2014.01.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2012] [Revised: 12/26/2013] [Accepted: 01/20/2014] [Indexed: 11/28/2022]
|
67
|
Badaoui B, Rutigliano T, Anselmo A, Vanhee M, Nauwynck H, Giuffra E, Botti S. RNA-sequence analysis of primary alveolar macrophages after in vitro infection with porcine reproductive and respiratory syndrome virus strains of differing virulence. PLoS One 2014; 9:e91918. [PMID: 24643046 PMCID: PMC3958415 DOI: 10.1371/journal.pone.0091918] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Accepted: 02/18/2014] [Indexed: 12/03/2022] Open
Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) mainly infects porcine alveolar macrophages (PAMs), resulting in porcine reproductive and respiratory syndrome (PRRS) in pigs. Most of the transcriptomic studies on PAMs infected with PRRSV conducted thus far have made use of microarray technology. Here, we investigated the transcriptome of PAMs in vitro at 12 h post-infection with two European PRRSV strains characterized by low (Lelystad, LV) and high (Lena) virulence through RNA-Seq. The expression levels of genes, isoforms, alternative transcription start sites (TSS) and differential promoter usage revealed a complex pattern of transcriptional and post-transcriptional gene regulation upon infection with the two strains. Gene ontology analysis confirmed that infection of PAMs with both the Lena and LV strains affected signaling pathways directly linked to the innate immune response, including interferon regulatory factors (IRF), RIG1-like receptors, TLRs and PKR pathways. The results confirmed that interferon signaling is crucial for transcriptional regulation during PAM infection. IFN-β1 and IFN-αω, but not IFN-α, were up-regulated following infection with either the LV or Lena strain. The down-regulation of canonical pathways, such as the interplay between the innate and adaptive immune responses, cell death and TLR3/TLR7 signaling, was observed for both strains, but Lena triggered a stronger down-regulation than LV. This analysis contributes to a better understanding of the interactions between PRRSV and PAMs and outlines the differences in the responses of PAMs to strains with different levels of virulence, which may lead to the development of new PRRSV control strategies.
Collapse
Affiliation(s)
- Bouabid Badaoui
- Parco Tecnologico Padano, Via Einstein, Lodi, Italy
- * E-mail:
| | | | - Anna Anselmo
- Parco Tecnologico Padano, Via Einstein, Lodi, Italy
| | - Merijn Vanhee
- Laboratory of Virology, Faculty of Veterinary Medicine, Ghent University, Ghent, Belgium
| | - Hans Nauwynck
- Laboratory of Virology, Faculty of Veterinary Medicine, Ghent University, Ghent, Belgium
| | | | - Sara Botti
- Parco Tecnologico Padano, Via Einstein, Lodi, Italy
| |
Collapse
|
68
|
Han D, Hu Y, Li L, Tian H, Chen Z, Wang L, Ma H, Yang H, Teng K. Highly pathogenic porcine reproductive and respiratory syndrome virus infection results in acute lung injury of the infected pigs. Vet Microbiol 2014; 169:135-46. [PMID: 24472226 PMCID: PMC7127595 DOI: 10.1016/j.vetmic.2013.12.022] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Revised: 12/19/2013] [Accepted: 12/23/2013] [Indexed: 01/11/2023]
Abstract
Highly pathogenic porcine reproductive and respiratory syndrome virus (HP-PRRSV) was firstly characterized in 2006 in China. The virus has caused great economic loss to the Chinese swine production during the past years. Herein, we experimentally infected SPF pigs using two strains of PRRSV with different pathogenicity and observed the lung pathological changes looking for new sights on the possible pathogenesis associated with the virulence of HP-PRRSV. The results indicated that the HP-PRRSV-infected pigs died and exhibited severe pathological changes of lungs featuring increased neutrophils, mast cells and mononuclear macrophages, compared with the pigs inoculated with low pathogenic (LP-) PRRSV. Furthermore, the pigs infected with HP-PRRSV showed the higher levels of tumor necrosis factor (TNF)-α, interleukin (IL)-1β, interleukin (IL)-8 and histamine, leukotriene B4 (LTB4), platelet activation factor (PAF) in sera than those inoculated with LP-PRRSV. Additionally, the fibrosis of lung was observed in the HP-PRRSV-infected pigs. At present, our findings suggest that the aberrant immune responses triggered by HP-PRRSV infection are closely related to acute lung injury (ALI), and especially the pathological changes in lung vascular system are of particular significance. These associated pathological changes of lung are in part responsible for the additional morbidity and mortality observed in HP-PRRSV infection.
Collapse
Affiliation(s)
- Deping Han
- Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine and Key state Laboratory of Agrobiotechnology, China Agricultural University, No.2 Yuanmingyuan West Road, Beijing, 100193, People's Republic of China
| | - Yanxin Hu
- Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine and Key state Laboratory of Agrobiotechnology, China Agricultural University, No.2 Yuanmingyuan West Road, Beijing, 100193, People's Republic of China
| | - Limin Li
- Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine and Key state Laboratory of Agrobiotechnology, China Agricultural University, No.2 Yuanmingyuan West Road, Beijing, 100193, People's Republic of China
| | - Haiyan Tian
- Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine and Key state Laboratory of Agrobiotechnology, China Agricultural University, No.2 Yuanmingyuan West Road, Beijing, 100193, People's Republic of China
| | - Zhi Chen
- Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine and Key state Laboratory of Agrobiotechnology, China Agricultural University, No.2 Yuanmingyuan West Road, Beijing, 100193, People's Republic of China
| | - Lin Wang
- Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine and Key state Laboratory of Agrobiotechnology, China Agricultural University, No.2 Yuanmingyuan West Road, Beijing, 100193, People's Republic of China
| | - Haiyan Ma
- Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine and Key state Laboratory of Agrobiotechnology, China Agricultural University, No.2 Yuanmingyuan West Road, Beijing, 100193, People's Republic of China
| | - Hanchun Yang
- Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine and Key state Laboratory of Agrobiotechnology, China Agricultural University, No.2 Yuanmingyuan West Road, Beijing, 100193, People's Republic of China.
| | - Kedao Teng
- Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine and Key state Laboratory of Agrobiotechnology, China Agricultural University, No.2 Yuanmingyuan West Road, Beijing, 100193, People's Republic of China.
| |
Collapse
|
69
|
García-Nicolás O, Baumann A, Vielle NJ, Gómez-Laguna J, Quereda JJ, Pallarés FJ, Ramis G, Carrasco L, Summerfield A. Virulence and genotype-associated infectivity of interferon-treated macrophages by porcine reproductive and respiratory syndrome viruses. Virus Res 2014; 179:204-11. [DOI: 10.1016/j.virusres.2013.08.009] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Revised: 08/21/2013] [Accepted: 08/22/2013] [Indexed: 10/26/2022]
|
70
|
Rodríguez-Gómez IM, Gómez-Laguna J, Carrasco L. Impact of PRRSV on activation and viability of antigen presenting cells. World J Virol 2013; 2:146-151. [PMID: 24286035 PMCID: PMC3832909 DOI: 10.5501/wjv.v2.i4.146] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Revised: 08/29/2013] [Accepted: 10/16/2013] [Indexed: 02/05/2023] Open
Abstract
Porcine reproductive and respiratory syndrome (PRRS) is one of the most important diseases of swine industry. The causal agent, PRRS-virus (PRRSV), is able to evade the host immune response and survive in the organism causing transient infections. Despite all scientific efforts, there are still some gaps in the knowledge of the pathogenesis of this disease. Antigen presenting cells (APCs), as initiators of the immune response, are located in the first line of defense against microorganisms, and are responsible for antigen recognition, processing and presentation. Dendritic cells (DCs) are the main type of APC involved in antigen presentation and they are susceptible to PRRSV infection. Thus, PRRSV replication in DCs may trigger off different mechanisms to impair the onset of a host effective immune response against the virus. On the one side, PRRSV may impair the basic functions of DCs by regulating the expression of major histocompatibility complex class II and CD80/86. Other strategy followed by the virus is the induction of cell death of APCs by apoptosis, necrosis or both of them. The impairment and/or cell death of APCs could lead to a failure in the onset of an efficient immune response, as long as cells could not properly activate T cells. Future aspects to take into account are also discussed in this review.
Collapse
|
71
|
Phenotypic modulation and cytokine profiles of antigen presenting cells by European subtype 1 and 3 porcine reproductive and respiratory syndrome virus strains in vitro and in vivo. Vet Microbiol 2013; 167:638-50. [PMID: 24120935 DOI: 10.1016/j.vetmic.2013.09.021] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Revised: 09/13/2013] [Accepted: 09/17/2013] [Indexed: 12/20/2022]
Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) causes continuous problems in the pig industry, due to high costs of outbreaks and reduced welfare of diseased pigs. The severity of infection is, partly, dependent on the virus strain. Recently isolated Eastern-European subtype 3 strains are more pathogenic than the widespread subtype 1 strains. There is, however, almost no information available about the mechanisms involved in the pathogenicity of these subtype 3 strains. The objective of the present study was to characterize the in vitro and in vivo response of two European subtype 1 strains, Belgium A and Lelystad-Ter Huurne (LV), and a virulent subtype 3 strain, Lena, in bone marrow-derived dendritic cells (BM-DC) (in vitro) and alveolar macrophages (in vitro and in vivo). It was shown that infection with the Lena strain resulted in a higher apoptosis of cells in vitro and a higher level of infectivity in vitro and in vivo than the other virus strains. Furthermore, infection with Lena resulted in a small downregulation of the immunologically relevant cell surface molecules SLA-I, SLA-II and CD80/86 in vitro, and SLA-II in vivo. In spite of these differences, in vitro cytokine responses did not differ significantly between strains, except for the absence of IL-10 production by Lena in BM-DC. The higher infectivity, apoptosis and downregulation of the cell surface molecules, may have contributed to the increased pathogenicity of Lena, and have dampened specific immune responses. This could explain the delayed and decreased adaptive immune responses observed after infections with this strain.
Collapse
|
72
|
Guo B, Lager KM, Schlink SN, Kehrli ME, Brockmeier SL, Miller LC, Swenson SL, Faaberg KS. Chinese and Vietnamese strains of HP-PRRSV cause different pathogenic outcomes in United States high health swine. Virology 2013; 446:238-50. [PMID: 24074587 DOI: 10.1016/j.virol.2013.08.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2013] [Revised: 06/24/2013] [Accepted: 08/09/2013] [Indexed: 01/12/2023]
Abstract
An infectious clone of a highly pathogenic PRRSV strain from Vietnam (rSRV07) was prepared and was demonstrated to contain multiple amino acid differences throughout the genome when compared to Chinese highly pathogenic PRRSV strain rJXwn06. Virus rescued from the rSRV07 infectious clone was compared to rJXwn06 and US Type 2 prototype strain VR-2332 to examine the effects of virus genotype and phenotype on in vitro growth, and virus challenge dose on in vivo pathogenicity and host response. After swine inoculation at high- and low-doses of virus, rSRV07 was shown to replicate to an approximately 10-fold lower level in serum than rJXwn06, produced lower body temperatures than rJXwn06 and resulted in decreased mortality. Furthermore, a 9-plex cytokine panel revealed that the cytokine responses varied between different strains of PRRSV, as well as between tissues examined and by inoculum dose.
Collapse
Affiliation(s)
- Baoqing Guo
- Veterinary Diagnostic & Production Animal Medicine, Iowa State University, Ames, IA, USA
| | | | | | | | | | | | | | | |
Collapse
|
73
|
Baumann A, Mateu E, Murtaugh MP, Summerfield A. Impact of genotype 1 and 2 of porcine reproductive and respiratory syndrome viruses on interferon-α responses by plasmacytoid dendritic cells. Vet Res 2013; 44:33. [PMID: 23675981 PMCID: PMC3672080 DOI: 10.1186/1297-9716-44-33] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Accepted: 04/18/2013] [Indexed: 12/31/2022] Open
Abstract
Porcine reproductive and respiratory syndrome (PRRS) virus (PRRSV) infections are characterized by prolonged viremia and viral shedding consistent with incomplete immunity. Type I interferons (IFN) are essential for mounting efficient antiviral innate and adaptive immune responses, but in a recent study, North American PRRSV genotype 2 isolates did not induce, or even strongly inhibited, IFN-α in plasmacytoid dendritic cells (pDC), representing “professional IFN-α-producing cells”. Since inhibition of IFN-α expression might initiate PRRSV pathogenesis, we further characterized PRRSV effects and host modifying factors on IFN-α responses of pDC. Surprisingly, a variety of type 1 and type 2 PRRSV directly stimulated IFN-α secretion by pDC. The effect did not require live virus and was mediated through the TLR7 pathway. Furthermore, both IFN-γ and IL-4 significantly enhanced the pDC production of IFN-α in response to PRRSV exposure. PRRSV inhibition of IFN-α responses from enriched pDC stimulated by CpG oligodeoxynucleotides was weak or absent. VR-2332, the prototype genotype 2 PRRSV, only suppressed the responses by 34%, and the highest level of suppression (51%) was induced by a Chinese highly pathogenic PRRSV isolate. Taken together, these findings demonstrate that pDC respond to PRRSV and suggest that suppressive activities on pDC, if any, are moderate and strain-dependent. Thus, pDC may be a source of systemic IFN-α responses reported in PRRSV-infected animals, further contributing to the puzzling immunopathogenesis of PRRS.
Collapse
Affiliation(s)
- Arnaud Baumann
- Institute of Virology and Immunoprophylaxis (IVI), Sensemattstrasse 293, Mittelhäusern, 3147, Switzerland.
| | | | | | | |
Collapse
|
74
|
Yang Q, Zhang Y, Chen J, Zhou Y, Li N, Qin Y, Yang M, Xia P, Cui B. Ligation of porcine Fc gamma receptor I inhibits levels of antiviral cytokine in response to PRRSV infection in vitro. Virus Res 2013; 173:421-5. [DOI: 10.1016/j.virusres.2013.02.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2012] [Revised: 02/02/2013] [Accepted: 02/04/2013] [Indexed: 11/17/2022]
|
75
|
Díaz I, Gimeno M, Callén A, Pujols J, López S, Charreyre C, Joisel F, Mateu E. Comparison of different vaccination schedules for sustaining the immune response against porcine reproductive and respiratory syndrome virus. Vet J 2013; 197:438-44. [PMID: 23499541 DOI: 10.1016/j.tvjl.2013.02.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Revised: 01/11/2013] [Accepted: 02/07/2013] [Indexed: 11/30/2022]
Abstract
In order to better understand how immunization against porcine reproductive and respiratory syndrome virus (PRRSV) can be improved using commercial vaccines, different strategies of immunization were applied in the field using an inactivated vaccine (INV), a modified live vaccine (MLV) or a combination of the two and the responses compared. In experiment 1 (E1), 21 piglets were distributed in three groups. Group A was vaccinated with a commercial INV at 2.5, 3.5 and 6.5 months old; group B pigs received the INV at 1.5, 2.5, 5.5 and 6.5 months old, while pigs in group C were kept as unvaccinated controls. At 7.5 months of age all pigs were challenged with PRRSV and followed for 21 days. In experiment 2 (E2), 32 piglets were distributed evenly in four groups. Groups A, B and C were vaccinated with a commercial MLV at 1.5 months old, while group D pigs were kept as controls. At 4.5 months old, groups A and C received the INV while B received a second MLV, 1 month later group C pigs received a third INV. At 6.5 months old all pigs were challenged as in E1. In both experiments, total antibodies, neutralizing antibodies (NA) and cell-mediated immunity (CMI) were evaluated, and viraemia was determined after challenge. In E1, immunization with an INV induced high interferon-γ responses after the second and subsequent vaccinations. Development of NA after challenge was faster in INV vaccinated pigs compared to unvaccinated controls. In E2, re-vaccination with INV induced NA responses similar to re-vaccination with MLV; however, a significant increase in NA titres after challenge was only detected in group C pigs. The use of combined protocols (MLV+INV) was superior to the use of MLV alone in inducing cell mediated immunity. In conclusion, the highest immune responses against PRRSV after a single shot were achieved with MLV; after that, INV re-vaccination should be considered as the best strategy to induce significant boosters.
Collapse
Affiliation(s)
- I Díaz
- Centre de Recerca en Sanitat Animal (CReSA), UAB-IRTA, Campus de la Universitat Autònoma de Barcelona, 08193 Bellaterra, Barcelona, Spain.
| | | | | | | | | | | | | | | |
Collapse
|
76
|
Gómez-Laguna J, Salguero FJ, Pallarés FJ, Carrasco L. Immunopathogenesis of porcine reproductive and respiratory syndrome in the respiratory tract of pigs. Vet J 2012; 195:148-55. [PMID: 23265866 PMCID: PMC7128372 DOI: 10.1016/j.tvjl.2012.11.012] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2012] [Revised: 11/09/2012] [Accepted: 11/15/2012] [Indexed: 12/12/2022]
Abstract
Porcine reproductive and respiratory syndrome (PRRS) virus (PRRSV) impairs local pulmonary immune responses by damaging the mucociliary transport system, impairing the function of porcine alveolar macrophages and inducing apoptosis of immune cells. An imbalance between pro- and anti-inflammatory cytokines, including tumour necrosis factor-α and interleukin-10, in PRRS may impair the immune response of the lung. Pulmonary macrophage subpopulations have a range of susceptibilities to different PRRSV strains and different capacities to express cytokines. Infection with PRRSV decreases the bactericidal activity of macrophages, which increases susceptibility to secondary bacterial infections. PRRSV infection is associated with an increase in concentrations of haptoglobin, which may interact with the virus receptor (CD163) and induce the synthesis of anti-inflammatory mediators. The balance between pro- and anti-inflammatory cytokines modulates the expression of CD163, which may affect the pathogenicity and replication of the virus in different tissues. With the emergence of highly pathogenic PRRSV, there is a need for more information on the immunopathogenesis of different strains of PRRS, particularly to develop more effective vaccines.
Collapse
Affiliation(s)
- Jaime Gómez-Laguna
- Research and Development R&D, Centro de Investigación y Calidad Agroalimentaria del Valle de los Pedroches CICAP, 14400 Pozoblanco, Córdoba, Spain.
| | | | | | | |
Collapse
|
77
|
Ferrari L, Martelli P, Saleri R, De Angelis E, Cavalli V, Bresaola M, Benetti M, Borghetti P. Lymphocyte activation as cytokine gene expression and secretion is related to the porcine reproductive and respiratory syndrome virus (PRRSV) isolate after in vitro homologous and heterologous recall of peripheral blood mononuclear cells (PBMC) from pigs vaccinated and exposed to natural infection. Vet Immunol Immunopathol 2012; 151:193-206. [PMID: 23228653 DOI: 10.1016/j.vetimm.2012.11.006] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2012] [Revised: 11/09/2012] [Accepted: 11/12/2012] [Indexed: 10/27/2022]
Abstract
The present study evaluated the lymphocyte activation in PRRSV-vaccinated pigs subsequently exposed to natural infection by in vitro stimulation of peripheral blood mononuclear cells (PBMC) with homologous vaccine and two heterologous PRRSV isolates. The responsiveness was assessed by determining IFN-γ secreting cells by ELISpot assay, lymphocyte CD8 phenotype by intracellular staining/flow cytometry, cytokine gene expression by real-time quantitative PCR and cytokine secretion by ELISA. Conventional pigs were weaned at 28 days of age and inoculated intramuscularly (IM) or needle-less intradermally (ID) with a modified-live PRRSV vaccine suspended in adjuvant, while control pigs were injected with adjuvant alone (ADJ). Blood samples were collected at vaccination, 35 days post-vaccination and after 35 days post-exposure to natural infection by a heterologous field strain. Thirty-five days post-vaccination, PRRSV vaccine induced a low but significant virus-specific IFN-γ secreting cell response upon stimulation with both the vaccine strain and the two isolates in vaccinated pigs. Conversely, after 35 days post-exposure, only the vaccine strain and the BS/114/S isolate triggered this response. Intracellular staining showed that PRRSV-specific immune cells reacting upon vaccine strain and BS/114/S stimulation were mostly CD8(+) IFN-γ producing cells whereas the stimulation with BS/55 isolate induced an IFN-γ production associated to the CD8(-)IFN-γ(+) phenotype. At 35 days post-vaccination, PBMC from vaccinated pigs showed lower IL-10 expression and release, and higher TNF-α gene expression upon stimulation with both the vaccine and viral isolates. After infection, both cytokines were not differently modulated in different groups. Immune parameters give evidence that IFN-γ secreting cells in the peripheral blood can be elicited upon PRRSV infection although vaccination itself does not stimulate high levels of these reactive cells. Moreover, the cross-reactivity against divergent PRRS viruses can show a different intensity and be differently associated with cytotoxic CD8(+)IFN-γ(+) as well as CD8(-)IFN-γ(+) cells. Overall, the obtained data confirmed that the immune activation against PRRSV is not dependent on the genetic divergence of the virus. Especially after infection, a different immune reactivity was evident upon stimulation with the different isolates in terms of frequency and CD8 phenotype of PRRSV-specific IFN-γ producing cells. The modulation of cytokines in vaccinated pigs appeared to be more dependent on vaccination or infection conditions than on stimulation by different isolates, and the changes of IL-10 more relevant than those of TNF-α at gene and protein levels. Moreover, under the conditions of this study, the PRRSV vaccine administered via the intradermal route by a needle-less device was confirmed to induce an immune response comparable or in some cases higher than the intramuscular route.
Collapse
Affiliation(s)
- Luca Ferrari
- Department of Veterinary Sciences, University of Parma, Via del Taglio, 10 - 43126 Parma, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
78
|
Barranco I, Gómez-Laguna J, Rodríguez-Gómez I, Quereda J, Salguero F, Pallarés F, Carrasco L. Immunohistochemical expression of IL-12, IL-10, IFN-α and IFN-γ in lymphoid organs of porcine reproductive and respiratory syndrome virus-infected pigs. Vet Immunol Immunopathol 2012; 149:262-71. [DOI: 10.1016/j.vetimm.2012.07.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2012] [Revised: 07/18/2012] [Accepted: 07/24/2012] [Indexed: 12/29/2022]
|
79
|
Rodríguez-Gómez IM, Gómez-Laguna J, Barranco I, Pallarés FJ, Ramis G, Salguero FJ, Carrasco L. Downregulation of antigen-presenting cells in tonsil and lymph nodes of porcine reproductive and respiratory syndrome virus-infected pigs. Transbound Emerg Dis 2012; 60:425-37. [PMID: 22816521 DOI: 10.1111/j.1865-1682.2012.01363.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) can persist in different organs of infected pigs, which suggests a failure in the immune response. Antigen-presenting cells (APCs) play a pivotal role in the induction of effective T- and B-cell responses. In this study, we investigated the changes in the different APC subpopulations and T- and B-cell counts in the tonsil, retropharyngeal and mediastinal lymph nodes of pigs experimentally infected with a European PRRSV field isolate. Our results demonstrated that the expression of S100, SWC3, HLA-DR molecule and CD3 was diminished in the studied organs throughout the study, observing a significant negative correlation between viral antigen and HLA-DR expression in both retropharyngeal and mediastinal lymph nodes. In contrast, λ-light chains showed an increase during the study. Taking all into account, after PRRSV infection, no enhancement in the number of APCs and T cells was observed, suggesting an impairment of the immune function which may allow the persistence of PRRSV into the organism.
Collapse
Affiliation(s)
- I M Rodríguez-Gómez
- Department of Anatomy and Comparative Pathology, Faculty of Veterinary Medicine, Córdoba University, Córdoba, Spain.
| | | | | | | | | | | | | |
Collapse
|
80
|
Zhang Y, Zhou Y, Yang Q, Mu C, Duan E, Chen J, Yang M, Xia P, Cui B. Ligation of Fc gamma receptor IIB enhances levels of antiviral cytokine in response to PRRSV infection in vitro. Vet Microbiol 2012; 160:473-80. [PMID: 22771209 DOI: 10.1016/j.vetmic.2012.06.021] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2012] [Accepted: 06/19/2012] [Indexed: 10/28/2022]
Abstract
PRRSV infection ADE facilitates the attachment and internalization of the virus onto its host cells, such as monocytes and macrophages, through Fc receptor-mediated endocytosis. FcγRIIB is the only inhibitory receptor with a tyrosine-based inhibitory motif (ITIM) in its cytoplasmic tail, where counters the "ITAM triggered" activation signals and down-regulates phagocytosis. However, porcine FcγRIIB's role in the antiviral immune response to PRRSV infection has not been studied. In this study, our results indicated that selective activation of porcine FcγRIIB in PAM cells up-regulated significantly mRNA levels of IFN-α and TNF-α at any time point post-pretreatment, suggesting that porcine FcγRIIB signal can enhance the innate antiviral response of host cells. PRRSV infection assay mediated by FcγRIIB indicated that selective activation of porcine FcγRIIB in PAM cells enhanced mRNA levels of antiviral cytokine (IFN-α and TNF-α) and repressed mRNA levels of IL-10 in response to PRRSV infection, suggesting that FcγRIIB ligation can enhance the antiviral immune response to PRRSV infection. In addition, FcγRIIB ligation to infection indicated that PRRSV replication in PAM was not positive correlation with increasing of IFN-α mRNA levels and decreasing of IL-10 mRNA levels, suggesting that there is complex viral replication mechanism in immune cells such as PAM for PRRSV.
Collapse
Affiliation(s)
- Yina Zhang
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou 450002, China
| | | | | | | | | | | | | | | | | |
Collapse
|
81
|
Subramaniam S, Beura LK, Kwon B, Pattnaik AK, Osorio FA. Amino acid residues in the non-structural protein 1 of porcine reproductive and respiratory syndrome virus involved in down-regulation of TNF-α expression in vitro and attenuation in vivo. Virology 2012; 432:241-9. [PMID: 22699004 DOI: 10.1016/j.virol.2012.05.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2012] [Revised: 05/03/2012] [Accepted: 05/14/2012] [Indexed: 10/28/2022]
Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) suppresses tumor necrosis factor-alpha (TNF-α) production at both transcriptional and post-transcriptional levels by its non-structural proteins 1α and 1β (Nsp1α and Nsp1β). To identify the amino acid residues responsible for this activity, we generated several alanine substitution mutants of Nsp1α and Nsp1β. Examination of the mutant proteins revealed that Nsp1α residues Gly90, Asn91, Arg97, Arg100 and Arg124 were necessary for TNF-α promoter suppression, whereas several amino acids spanning the entire Nsp1β were found to be required for this activity. Two mutant viruses, with mutations at Nsp1α Gly90 or Nsp1β residues 70-74, generated from infectious cDNA clones, exhibited attenuated viral replication in vitro and TNF-α was found to be up regulated in infected macrophages. In infected pigs, the Nsp1β mutant virus was attenuated in growth. These studies provide insights into how PRRSV evades the effector mechanisms of innate immunity during infection.
Collapse
Affiliation(s)
- Sakthivel Subramaniam
- School of Veterinary Medicine & Biomedical Sciences and Nebraska Center for Virology, University of Nebraska-Lincoln, NE 68583, USA
| | | | | | | | | |
Collapse
|
82
|
Regulatory T cells in arterivirus and coronavirus infections: do they protect against disease or enhance it? Viruses 2012; 4:833-46. [PMID: 22754651 PMCID: PMC3386620 DOI: 10.3390/v4050833] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2012] [Revised: 05/03/2012] [Accepted: 05/07/2012] [Indexed: 01/05/2023] Open
Abstract
Regulatory T cells (Tregs) are a subset of T cells that are responsible for maintaining peripheral immune tolerance and homeostasis. The hallmark of Tregs is the expression of the forkhead box P3 (FoxP3) transcription factor. Natural regulatory T cells (nTregs) are a distinct population of T cells that express CD4 and FoxP3. nTregs develop in the thymus and function in maintaining peripheral immune tolerance. Other CD4+, CD4-CD8-, and CD8+CD28- T cells can be induced to acquire regulatory function by antigenic stimulation, depending on the cytokine milieu. Inducible (or adaptive) Tregs frequently express high levels of the interleukin 2 receptor (CD25). Atypical Tregs express FoxP3 and CD4 but have no surface expression of CD25. Type 1 regulatory T cells (Tr1 cells) produce IL-10, while T helper 3 cells (Th3) produce TGF-β. The function of inducible Tregs is presumably to maintain immune homeostasis, especially in the context of chronic inflammation or infection. Induction of Tregs in coronaviral infections protects against the more severe forms of the disease attributable to the host response. However, arteriviruses have exploited these T cell subsets as a means to dampen the immune response allowing for viral persistence. Treg induction or activation in the pathogenesis of disease has been described in both porcine reproductive and respiratory syndrome virus, lactate dehydrogenase elevating virus, and mouse hepatitis virus. This review discusses the development and biology of regulatory T cells in the context of arteriviral and coronaviral infection.
Collapse
|
83
|
Cecere TE, Meng XJ, Pelzer K, Todd SM, Beach NM, Ni YY, Leroith T. Co-infection of porcine dendritic cells with porcine circovirus type 2a (PCV2a) and genotype II porcine reproductive and respiratory syndrome virus (PRRSV) induces CD4(+)CD25(+)FoxP3(+) T cells in vitro. Vet Microbiol 2012; 160:233-9. [PMID: 22633482 PMCID: PMC3443269 DOI: 10.1016/j.vetmic.2012.04.040] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2011] [Revised: 04/23/2012] [Accepted: 04/30/2012] [Indexed: 12/05/2022]
Abstract
Porcine circovirus associated disease (PCVAD) is currently one of the most economically important diseases in the global swine industry. Porcine circovirus type 2 (PCV2) is the primary causative agent, however co-infection with other swine pathogens such as porcine reproductive and respiratory syndrome virus (PRRSV) is often required to induce the full spectrum of clinical PCVAD. While the specific mechanisms of viral co-infection that lead to clinical disease are not fully understood, immune modulation by the co-infecting viruses likely plays a critical role. We evaluated the ability of dendritic cells (DC) infected with PRRSV, PCV2, or both to induce regulatory T cells (Tregs) in vitro. DCs infected with PCV2 significantly increased CD4+CD25+FoxP3+ Tregs (p < 0.05) and DCs co-infected with PRRSV and PCV2 induced significantly higher numbers of Tregs than with PCV2 alone (p < 0.05). Cytokine analysis indicated that the induction of Tregs by co-infected DCs may be dependent on TGF-β and not IL-10. Our data support the immunomodulatory role of PCV2/PRRSV co-infection in the pathogenesis of PCVAD, specifically via Treg-mediated immunosuppression.
Collapse
Affiliation(s)
- T E Cecere
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland Regional College of Veterinary Medicine, Virginia Tech, Blacksburg, VA 24061, USA.
| | | | | | | | | | | | | |
Collapse
|
84
|
Díaz I, Gimeno M, Darwich L, Navarro N, Kuzemtseva L, López S, Galindo I, Segalés J, Martín M, Pujols J, Mateu E. Characterization of homologous and heterologous adaptive immune responses in porcine reproductive and respiratory syndrome virus infection. Vet Res 2012; 43:30. [PMID: 22515169 PMCID: PMC3403850 DOI: 10.1186/1297-9716-43-30] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2011] [Accepted: 04/19/2012] [Indexed: 11/24/2022] Open
Abstract
The present study characterized the homologous and heterologous immune response in type-I porcine reproductive and respiratory syndrome virus (PRRSV) infection. Two experiments were conducted: in experiment 1, eight pigs were inoculated with PRRSV strain 3262 and 84 days post-inoculation (dpi) they were challenged with either strain 3262 or strain 3267 and followed for the next 14 days (98 dpi). In experiment 2, eight pigs were inoculated with strain 3267 and challenged at 84 dpi as above. Clinical course, viremia, humoral response (neutralizing and non-neutralizing antibodies, NA) and virus-specific IFN-γ responses (ELISPOT) were evaluated all throughout the study. Serum levels of IL-1, IL-6, IL-8, TNF-α and TGF-β were determined (ELISA) after the second challenge. In experiment 1 primo-inoculation with strain 3262 induced viremia of ≤ 28 days, low titres of homologous NA but strong IFN-γ responses. In contrast, strain 3267 induced longer viremias (up to 56 days), higher NA titres (≤ 6 log2) and lower IFN-γ responses. Inoculation with 3267 produced higher serum IL-8 levels. After the re-challenge at 84 dpi, pigs in experiment 1 developed mostly a one week viremia regardless of the strain used. In experiment 2, neither the homologous nor the heterologous challenge resulted in detectable viremia although PRRSV was present in tonsils of some animals. Homologous re-inoculation with 3267 produced elevated TGF-β levels in serum for 7–14 days but this did not occur with the heterologous re-inoculation. In conclusion, inoculation with different PRRSV strains result in different virological and immunological outcomes and in different degrees of homologous and heterologous protection.
Collapse
Affiliation(s)
- Ivan Díaz
- Centre de Recerca en Sanitat Animal (CReSA), UAB-IRTA, Campus de la Universitat Autònoma de Barcelona, 08193, Bellaterra, Barcelona, Spain.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
85
|
Hou J, Wang L, He W, Zhang H, Feng WH. Highly pathogenic porcine reproductive and respiratory syndrome virus impairs LPS- and poly(I:C)-stimulated tumor necrosis factor-alpha release by inhibiting ERK signaling pathway. Virus Res 2012; 167:106-11. [PMID: 22497732 DOI: 10.1016/j.virusres.2012.03.017] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2012] [Revised: 03/25/2012] [Accepted: 03/27/2012] [Indexed: 11/29/2022]
Abstract
Atypical porcine reproductive and respiratory syndrome (PRRS) characterized by high morbidity and mortality emerged in China in 2006. The causative agent was confirmed to be a highly pathogenic PRRS virus (HP-PRRSV). However, the pathogenesis of HP-PRRSV is still uncertain. Here, the ability of the highly pathogenic strains (HV and JX) to induce tumor necrosis factor alpha (TNF-α) was studied. Our results showed that HV and JX were weaker inducers of TNF-α than the conventional strain CH-1a. Moreover, HV infection was demonstrated to suppress extracellular signal-regulated kinase (ERK) phosphorylation at the early time points. Pharmacologic inhibition or activation of ERK revealed that TNF-α production in HV-infected macrophages was associated with the activation status of ERK. Furthermore, HV- and JX-infection could potently impair lipopolysaccharide (LPS)- and poly(I:C)-stimulated TNF-α release in a dose dependent manner whereas synergistic effects were observed at mRNA level. The observation suggested the involvement of posttranslational impact of HP-PRRSV on TNF-α production, which might be attributed to the reduced ERK1/2 phosphorylation in response to toll-like receptor (TLR)-ligation. Taken together, our results indicated that HP-PRRSV infection could impair TNF-α production by inhibiting ERK signaling pathway, which might partially contribute to the pathogenesis of HP-PRRSV.
Collapse
Affiliation(s)
- Jun Hou
- State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing 100193, China
| | | | | | | | | |
Collapse
|
86
|
Darwich L, Gimeno M, Sibila M, Diaz I, de la Torre E, Dotti S, Kuzemtseva L, Martin M, Pujols J, Mateu E. Genetic and immunobiological diversities of porcine reproductive and respiratory syndrome genotype I strains. Vet Microbiol 2011; 150:49-62. [PMID: 21310555 DOI: 10.1016/j.vetmic.2011.01.008] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2010] [Revised: 12/30/2010] [Accepted: 01/10/2011] [Indexed: 12/21/2022]
Abstract
Genetic diversity of porcine reproductive and respiratory syndrome virus (PRRSV) has been based on ORF5/GP5 and ORF7/N protein variations. Complete viral genome studies are limited and focused on a single or a few set of strains. Moreover, there is a general tendency to extrapolate results obtained from a single isolate to the overall PRRSV population. In the present study, six genotype-I isolates of PRRSV were sequenced from ORF1a to ORF7. Phylogenetic comparisons and the variability degree of known linear B-epitopes were done considering other available full-length genotype-I sequences. Cytokine induction of all strains was also evaluated in different cellular systems. Non structural protein 2 (nsp2) was the most variable part of the virus with 2 out of 6 strains harboring a 74 aa deletion. Deletions were also found in ORF3 and ORF4. Phylogenetic analyses showed that isolates could be grouped differently depending on the ORF examined and the highest similarity with the full genome cluster was found for the nsp9. Interestingly, most of predicted linear B-epitopes in the literature, particularly in nsp2 and GP4 regions, were found deleted or varied in some of our isolates. Moreover, 4 strains, those with deletions in nsp2, induced TNF-α and 3 induced IL-10. These results underline the high genetic diversity of PRRSV mainly in nsp1, nsp2 and ORFs 3 and 4. This variability also affects most of the known linear B-epitopes of the virus. Accordingly, different PRRSV strains might have substantially different immunobiological properties. These data can contribute to the understanding of PRRSV complexity.
Collapse
Affiliation(s)
- Laila Darwich
- Departament de Sanitat i Anatomia Animals, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain.
| | | | | | | | | | | | | | | | | | | |
Collapse
|