DOI QR코드

DOI QR Code

A standardized method to study immune responses using porcine whole blood

  • Sameer-ul-Salam Mattoo (Korea Zoonosis Research Institute) ;
  • Ram Prasad Aganja (College of Veterinary Medicine, Jeonbuk National University) ;
  • Seung-Chai Kim (College of Veterinary Medicine, Jeonbuk National University) ;
  • Chang-Gi Jeong (College of Veterinary Medicine, Jeonbuk National University) ;
  • Salik Nazki (The Pirbright Institute) ;
  • Amina Khatun (Department of Veterinary Pathology, Faculty of Animal Science and Veterinary Medicine, Sher-e-Bangla Agricultural University) ;
  • Won-Il Kim (College of Veterinary Medicine, Jeonbuk National University) ;
  • Sang-Myeong Lee (College of Veterinary Medicine, Chungbuk National University)
  • Received : 2022.08.20
  • Accepted : 2022.11.08
  • Published : 2023.01.31

Abstract

Background: Peripheral blood mononuclear cells (PBMCs) are commonly used to assess in vitro immune responses. However, PBMC isolation is a time-consuming procedure, introduces technical variability, and requires a relatively large volume of blood. By contrast, whole blood assay (WBA) is faster, cheaper, maintains more physiological conditions, and requires less sample volume, laboratory training, and equipment. Objectives: Herein, this study aimed to develop a porcine WBA for in vitro evaluation of immune responses. Methods: Heparinized whole blood (WB) was diluted (non-diluted, 1/2, 1/8, and 1/16) in RPMI-1640 media, followed by phorbol myristate acetate and ionomycin. After 24 h, cells were stained for interferon (IFN)-γ secreting T-cells followed by flow cytometry, and the supernatant was analyzed for tumor necrosis factor (TNF)-α. In addition, diluted WB was stimulated by lipopolysaccharide (LPS) and polyinosinic:polycytidylic acid (poly I:C), reference strain KCTC3557 (RS), field isolate (FI), of heat-killed (HK) Streptococcus suis, and porcine reproductive and respiratory syndrome virus (PRRSV). Results: The frequency of IFN-γ+CD3+ T-cells and concentration of TNF-α in the supernatant of WB increased with increasing dilution factor and were optimal at 1/8. WB TNF-α and interleukin (IL)-10 cytokine levels increased significantly following stimulation with LPS or poly I:C. Further, FI and RS induced IL-10 production in WB. Additionally, PRRSV strains increased the frequency of IFN-γ+ CD4-CD8+ cells, and IFN-γ was non-significantly induced in the supernatant of re-stimulated samples. Conclusions: We propose that the WBA is a rapid, reliable, and simple method to evaluate immune responses and WB should be diluted to trigger immune cells.

Keywords

Acknowledgement

This research was funded by a grant from "BioGreen21 Agri-Tech Innovation Program (Award number: PJ01561102, Recipient: Won-Il Kim)" of Rural Development Administration, the Ministry of Agriculture, Food and Rural Affairs of Korea and National Research Foundation of Korea (NRF) grant (Award number: 2019R1A2C2010432, Recipient: Sang-Myeong Lee) funded by the Korea Government (MIST).

References

  1. Haller D, Blum S, Bode C, Hammes WP, Schiffrin EJ. Activation of human peripheral blood mononuclear cells by nonpathogenic bacteria in vitro: evidence of NK cells as primary targets. Infect Immun. 2000;68(2):752-759. https://doi.org/10.1128/IAI.68.2.752-759.2000
  2. Sela U, Euler CW, Correa da Rosa J, Fischetti VA. Strains of bacterial species induce a greatly varied acute adaptive immune response: the contribution of the accessory genome. PLoS Pathog. 2018;14(1):e1006726.
  3. Gerner W, Kaser T, Pintaric M, Groiss S, Saalmuller A. Detection of intracellular antigens in porcine PBMC by flow cytometry: a comparison of fixation and permeabilisation reagents. Vet Immunol Immunopathol. 2008;121(3-4):251-259. https://doi.org/10.1016/j.vetimm.2007.09.019
  4. Shabir N, Khatun A, Nazki S, Gu S, Lee SM, Hur TY, et al. In vitro immune responses of porcine alveolar macrophages reflect host immune responses against porcine reproductive and respiratory syndrome viruses. BMC Vet Res. 2018;14(1):380. https://doi.org/10.1186/s12917-018-1686-7
  5. Gaines H, Andersson L, Biberfeld G. A new method for measuring lymphoproliferation at the single-cell level in whole blood cultures by flow cytometry. J Immunol Methods. 1996;195(1-2):63-72. https://doi.org/10.1016/0022-1759(96)00085-3
  6. Godoy-Ramirez K, Franck K, Mahdavifar S, Andersson L, Gaines H. Optimum culture conditions for specific and nonspecific activation of whole blood and PBMC for intracellular cytokine assessment by flow cytometry. J Immunol Methods. 2004;292(1-2):1-15. https://doi.org/10.1016/j.jim.2004.04.028
  7. Damsgaard CT, Lauritzen L, Calder PC, Kjaer TM, Frokiaer H. Whole-blood culture is a valid low-cost method to measure monocytic cytokines - a comparison of cytokine production in cultures of human whole-blood, mononuclear cells and monocytes. J Immunol Methods. 2009;340(2):95-101. https://doi.org/10.1016/j.jim.2008.10.005
  8. Hodge G, Hodge S, Han P. Increased levels of apoptosis of leukocyte subsets in cultured PBMCs compared to whole blood as shown by Annexin V binding: relevance to cytokine production. Cytokine. 2000;12(12):1763-1768. https://doi.org/10.1006/cyto.2000.0790
  9. Eskola J, Soppi E, Viljanen M, Ruuskanen O. A new micromethod for lymphocyte stimulation using whole blood. Immunol Commun. 1975;4(4):297-307. https://doi.org/10.3109/08820137509055781
  10. Digel W, Marcucci F, Kirchner H. Induction of interferon gamma in leucocyte cultures of the peripheral blood of mice. J Interferon Res. 1983;3(1):65-69. https://doi.org/10.1089/jir.1983.3.65
  11. Duffy D, Rouilly V, Libri V, Hasan M, Beitz B, David M, et al. Functional analysis via standardized whole-blood stimulation systems defines the boundaries of a healthy immune response to complex stimuli. Immunity. 2014;40(3):436-450. https://doi.org/10.1016/j.immuni.2014.03.002
  12. Santin M, Munoz L, Rigau D. Interferon-γ release assays for the diagnosis of tuberculosis and tuberculosis infection in HIV-infected adults: a systematic review and meta-analysis. PLoS One. 2012;7(3):e32482.
  13. Yancy H, Ayers SL, Farrell DE, Day A, Myers MJ. Differential cytokine mRNA expression in swine whole blood and peripheral blood mononuclear cell cultures. Vet Immunol Immunopathol. 2001;79(1-2):41-52. https://doi.org/10.1016/S0165-2427(01)00247-1
  14. Segura M, Vanier G, Al-Numani D, Lacouture S, Olivier M, Gottschalk M. Proinflammatory cytokine and chemokine modulation by Streptococcus suis in a whole-blood culture system. FEMS Immunol Med Microbiol. 2006;47(1):92-106. https://doi.org/10.1111/j.1574-695X.2006.00067.x
  15. Costers S, Lefebvre DJ, Goddeeris B, Delputte PL, Nauwynck HJ. Functional impairment of PRRSVspecific peripheral CD3+ CD8high cells. Vet Res. 2009;40(5):46.
  16. Nazki S, Khatun A, Jeong CG, Mattoo SU, Gu S, Lee SI, et al. Evaluation of local and systemic immune responses in pigs experimentally challenged with porcine reproductive and respiratory syndrome virus. Vet Res. 2020;51(1):66.
  17. Yancy H, Ayers SL, Farrell DE, Day A, Myers MJ. Differential cytokine mRNA expression in swine whole blood and peripheral blood mononuclear cell cultures. Vet Immunol Immunopathol. 2001;79(1-2):41-52. https://doi.org/10.1016/S0165-2427(01)00247-1
  18. May JE, Pemberton RM, Hart JP, McLeod J, Wilcock G, Doran O. Use of whole blood for analysis of disease-associated biomarkers. Anal Biochem. 2013;437(1):59-61. https://doi.org/10.1016/j.ab.2013.02.024
  19. Albers R, Antoine JM, Bourdet-Sicard R, Calder PC, Gleeson M, Lesourd B, et al. Markers to measure immunomodulation in human nutrition intervention studies. Br J Nutr. 2005;94(3):452-481. https://doi.org/10.1079/BJN20051469
  20. Appay V, Rowland-Jones SL. The assessment of antigen-specific CD8+ T cells through the combination of MHC class I tetramer and intracellular staining. J Immunol Methods. 2002;268(1):9-19. https://doi.org/10.1016/S0022-1759(02)00195-3
  21. Ai W, Li H, Song N, Li L, Chen H. Optimal method to stimulate cytokine production and its use in immunotoxicity assessment. Int J Environ Res Public Health. 2013;10(9):3834-3842. https://doi.org/10.3390/ijerph10093834
  22. Mascher B, Schlenke P, Seyfarth M. Expression and kinetics of cytokines determined by intracellular staining using flow cytometry. J Immunol Methods. 1999;223(1):115-121. https://doi.org/10.1016/S0022-1759(98)00200-2
  23. Olsnes C, Stavang H, Olofsson J, Aarstad HJ. TNF-α is secreted by monocytes in transit to become macrophages, but not by peripheral blood monocytes, following OK-432 (lyophilized S. pyogenes) stimulation. Scand J Immunol. 2007;66(6):684-693. https://doi.org/10.1111/j.1365-3083.2007.02016.x
  24. Baran J, Kowalczyk D, Ozog M, Zembala M. Three-color flow cytometry detection of intracellular cytokines in peripheral blood mononuclear cells: comparative analysis of phorbol myristate acetateionomycin and phytohemagglutinin stimulation. Clin Diagn Lab Immunol. 2001;8(2):303-313. https://doi.org/10.1128/CDLI.8.2.303-313.2001
  25. Dubravec DB, Spriggs DR, Mannick JA, Rodrick ML. Circulating human peripheral blood granulocytes synthesize and secrete tumor necrosis factor alpha. Proc Natl Acad Sci U S A. 1990;87(17):6758-6761. https://doi.org/10.1073/pnas.87.17.6758
  26. Leroux M, Schindler L, Braun R, Doerr HW, Geisen HP, Kirchner H. A whole-blood lymphoproliferation assay for measuring cellular immunity against herpes viruses. J Immunol Methods. 1985;79(2):251-262. https://doi.org/10.1016/0022-1759(85)90105-X
  27. Weir RE, Morgan AR, Britton WJ, Butlin CR, Dockrell HM. Development of a whole blood assay to measure T cell responses to leprosy: a new tool for immuno-epidemiological field studies of leprosy immunity. J Immunol Methods. 1994;176(1):93-101. https://doi.org/10.1016/0022-1759(94)90353-0
  28. Russell M, Mellkvist-Roos A, Mo J, Hidi R. Simple and robust two-step ex vivo whole blood stimulation assay suitable for investigating IL-17 pathway in a clinical laboratory setting. J Immunol Methods. 2018;454:71-75. https://doi.org/10.1016/j.jim.2017.11.001
  29. Ngkelo A, Meja K, Yeadon M, Adcock I, Kirkham PA. LPS induced inflammatory responses in human peripheral blood mononuclear cells is mediated through NOX4 and Giα dependent PI-3kinase signalling. J Inflamm (Lond). 2012;9(1):1-7. https://doi.org/10.1186/1476-9255-9-1
  30. Jansky L, Reymanova P, Kopecky J. Dynamics of cytokine production in human peripheral blood mononuclear cells stimulated by LPS or infected by Borrelia. Physiol Res. 2003;52(5):593-598. https://doi.org/10.33549/physiolres.930372
  31. Chen J, Bruns AH, Donnelly HK, Wunderink RG. Comparative in vitro stimulation with lipopolysaccharide to study TNFalpha gene expression in fresh whole blood, fresh and frozen peripheral blood mononuclear cells. J Immunol Methods. 2010;357(1-2):33-37. https://doi.org/10.1016/j.jim.2010.03.006
  32. Tobias PS, Soldau K, Ulevitch RJ. Isolation of a lipopolysaccharide-binding acute phase reactant from rabbit serum. J Exp Med. 1986;164(3):777-793. https://doi.org/10.1084/jem.164.3.777
  33. Wright SD, Ramos RA, Tobias PS, Ulevitch RJ, Mathison JC. CD14, a receptor for complexes of lipopolysaccharide (LPS) and LPS binding protein. Science. 1990;249(4975):1431-1433. https://doi.org/10.1126/science.1698311
  34. Dorresteijn MJ, Draisma A, van der Hoeven JG, Pickkers P. Lipopolysaccharide-stimulated whole blood cytokine production does not predict the inflammatory response in human endotoxemia. Innate Immun. 2010;16(4):248-253. https://doi.org/10.1177/1753425909339923
  35. Pjanova D, Mandrika L, Petrovska R, Vaivode K, Donina S. Comparison of the effects of bacteriophage-derived dsRNA and poly(I:C) on ex vivo cultivated peripheral blood mononuclear cells. Immunol Lett. 2019;212:114-119. https://doi.org/10.1016/j.imlet.2019.06.010
  36. Wang J, Wang Y, Wang H, Hao X, Wu Y, Guo J. Selection of reference genes for gene expression studies in porcine whole blood and peripheral blood mononuclear cells under polyinosinic: polycytidylic acid stimulation. Asian-Australas J Anim Sci. 2014;27(4):471-478. https://doi.org/10.5713/ajas.2013.13471
  37. Segura M, Stankova J, Gottschalk M. Heat-killed Streptococcus suis capsular type 2 strains stimulate tumor necrosis factor alpha and interleukin-6 production by murine macrophages. Infect Immun. 1999;67(9):4646-4654. https://doi.org/10.1128/iai.67.9.4646-4654.1999
  38. Diaz I, Darwich L, Pappaterra G, Pujols J, Mateu E. Different European-type vaccines against porcine reproductive and respiratory syndrome virus have different immunological properties and confer different protection to pigs. Virology. 2006;351(2):249-259. https://doi.org/10.1016/j.virol.2006.03.046
  39. Madapong A, Saeng-Chuto K, Boonsoongnern A, Tantituvanont A, Nilubol D. Cell-mediated immune response and protective efficacy of porcine reproductive and respiratory syndrome virus modified-live vaccines against co-challenge with PRRSV-1 and PRRSV-2. Sci Rep. 2020;10(1):1649.
  40. Liu Y, Che TM, Song M, Lee JJ, Almeida JA, Bravo D, et al. Dietary plant extracts improve immune responses and growth efficiency of pigs experimentally infected with porcine reproductive and respiratory syndrome virus. J Anim Sci. 2013;91(12):5668-5679. https://doi.org/10.2527/jas.2013-6495