Skip to main navigation menu Skip to main content Skip to site footer

Original article

Vol. 148 No. 3940 (2018)

The immune influence of a parabiosis model on tumour-bearing mice

  • Nan Feng
  • Jianmin Luo
  • Ximin Guo
DOI
https://doi.org/10.4414/smw.2018.14678
Cite this as:
Swiss Med Wkly. 2018;148:w14678
Published
03.10.2018

Summary

AIM

The aim of this study was to analyse the immune influence of a parabiosis model on tumour-bearing mice.

METHODS

Parabiosis was established between C57BL/6 wild-type mice expressing green fluorescent protein (GFP+) and C57BL/6 wild-type mice without green fluorescent protein (GFP−) to ensure blood cross-circulation between animals, and then the expression of CD4+ T cells, CD8+ T cells and interleukins 2, 4 and 10, and interferon-gamma (INF–γ) in spleen cells of parabiosis model mice were examined with flow cytometry. At day 8 and day 14 after conjoined surgery, we were aiming to sample tumour tissue in the parabiosis mice and observe changes of CD3, CD4, CD8, CD31, IFN-γ and vascular endothelial growth factor (VEGF) through immunohistochemical analysis.

RESULTS

The interaction of blood was established on the third day with modelling rate of 85.7% after blood interaction. The healthy cells of GFP+ C57 mice entered the blood circulation of tumour-bearing mice via a connecting capillary network, playing a role in stimulating CD4+ and CD8+ cells in the tumour-bearing mice so that CD4+ cells increased more in tumour-bearing mice than in the positive control group (p <0.05). The number of GFP+ cells that were detected in a tumour-bearing mouse was small, but GFP+ cells can stimulate the mouse itself to generate more CD4+/interleukin (IL)-4, CD4+/IL-10 (p <0.05).The numbers of CD4+/IL-2, CD4+/IL-4 and CD4+/IL-10 among the GFP+ mice were higher than those in the negative control group(p <0.05).The levels of IFN-γ in both mice in the parabiosis model were decreased (p <0.05). The rate of CD4+/CD8+ in parabiosis GFP+ mice was higher than in the negative control group (p <0.05). In immunohistochemical tests, the rates of CD3, CD4, CD8 and IFN-γ positive cells was higher than in the positive control group, with their optical densities of 0.32 ± 0.63, 0.33 ± 0.00, 0.31 ± 0.91 and 0.28 ± 0.14 respectively (p <0.05). The expression of CD31 (0.19 ± 0.50) and VEGF (0.19 ± 0.21) were lower when compared with the positive control group, with no significant difference. CD31 and VEFG cell expression was low, at 0.19 ± 0.50 and 0.19 ± 0.21, respectively, compared with the positive control group (p >0.05). Values for CD31 and VEGF cells in the positive control group were higher, at 0.32 ± 0.35 and 0.29 ± 0.35, respectively, but when compared with the parabiosis tumour-bearing group, there was no significant difference. The expression of CD3, CD4, CD8 and IFN-γ cells at day 8 was low: 0.22, 0.17, 0.15 and 0.16, respectively. When compared with the parabiosis tumour-bearing group, there was no significant difference.

CONCLUSIONS

The established allogeneic parabiosis mice model can be well adapted to the conjoined state of mice and be applied in wide medical experiments. The parabiosis model has played an important role in studying immune regulation, which provides a basis for the future tumour immunotherapy. Parabiosis models can stimulate tumour-bearing mice to generate CD3, CD4, CD8 and IFN-γ, and play a notable role in immune regulation and tumour destruction. The positive expression rates of CD31 and VEFG cells in the parabiosis tumour-bearing group were lower; however, when compared with the positive control group, there was no significant difference.

References

  1. Conboy IM, Conboy MJ, Wagers AJ, Girma ER, Weissman IL, Rando TA. Rejuvenation of aged progenitor cells by exposure to a young systemic environment. Nature. 2005;433(7027):760–4. doi:.https://doi.org/10.1038/nature03260
  2. Villeda SA, Luo J, Mosher KI, Zou B, Britschgi M, Bieri G, et al. The ageing systemic milieu negatively regulates neurogenesis and cognitive function. Nature. 2011;477(7362):90–4. doi:.https://doi.org/10.1038/nature10357
  3. Loffredo FS, Steinhauser ML, Jay SM, Gannon J, Pancoast JR, Yalamanchi P, et al. Growth differentiation factor 11 is a circulating factor that reverses age-related cardiac hypertrophy. Cell. 2013;153(4):828–39. doi:.https://doi.org/10.1016/j.cell.2013.04.015
  4. Castellano JM, Palner M, Li S-B, Freeman GM, Jr, Nguyen A, Shen B, et al. In vivo assessment of behavioral recovery and circulatory exchange in the peritoneal parabiosis model. Sci Rep. 2016;6:29015. doi:.https://doi.org/10.1038/srep29015
  5. Mittal D, Gubin MM, Schreiber RD, Smyth MJ. New insights into cancer immunoediting and its three component phases--elimination, equilibrium and escape. Curr Opin Immunol. 2014;27(27):16–25. doi:.https://doi.org/10.1016/j.coi.2014.01.004
  6. Li X, Du W, Liu W, Li X, Li H, Huang SA. Comprehensive flow cytometry phenotype in acute leukemia at diagnosis and at relapse. APMIS. 2010;118(5):353–9. doi:.https://doi.org/10.1111/j.1600-0463.2010.02603.x
  7. Zhang K, Deng H, Cagle PT. Utility of immunohistochemistry in the diagnosis of pleuropulmonary and mediastinal cancers: a review and update. Arch Pathol Lab Med. 2014;138(12):1611–28. doi:.https://doi.org/10.5858/arpa.2014-0092-RA
  8. Decouvelaere AV. Apport de l’immuno-histochimie dans le diagnostic des sarcomes [Immunohistochemistry in the diagnosis of sarcomas]. Ann Pathol. 2015;35(1):98–106. doi:.https://doi.org/10.1016/j.annpat.2014.11.006
  9. Eggel A, Wyss-Coray T. A revival of parabiosis in biomedical research. Swiss Med Wkly. 2014;144:w13914. doi:.https://doi.org/10.4414/smw.2014.13914
  10. Lunsford WR, McCAY CM, Lupien PJ, Pope FE, Sperling G. Parabiosis as a method for studying factors which affect aging in rats. Gerontologia. 1963;7(1):1–8. doi:.https://doi.org/10.1159/000211170
  11. Deacon RM. Assessing nest building in mice. Nat Protoc. 2006;1(3):1117–9. doi:.https://doi.org/10.1038/nprot.2006.170
  12. Kamran P, Sereti KI, Zhao P, Ali SR, Weissman IL, Ardehali R. Parabiosis in mice: a detailed protocol. J Vis Exp. 2013;80:50556.
  13. Rando TA, Chang HY. Aging, rejuvenation, and epigenetic reprogramming: resetting the aging clock. Cell. 2012;148(1-2):46–57. doi:.https://doi.org/10.1016/j.cell.2012.01.003
  14. Duyverman AM, Kohno M, Duda DG, Jain RK, Fukumura D. A transient parabiosis skin transplantation model in mice. Nat Protoc. 2012;7(4):763–70. doi:.https://doi.org/10.1038/nprot.2012.032
  15. Gaber T, Strehl C, Sawitzki B, Hoff P, Buttgereit F. Cellular energy metabolism in T-lymphocytes. Int Rev Immunol. 2015;34(1):34–49. doi:.https://doi.org/10.3109/08830185.2014.956358
  16. Dumitriu IE. The life (and death) of CD4+ CD28(null) T cells in inflammatory diseases. Immunology. 2015;146(2):185–93. doi:.https://doi.org/10.1111/imm.12506
  17. Plain KM, Verma ND, Tran GT, Nomura M, Boyd R, Robinson CM, et al. Cytokines affecting CD4(+) T regulatory cells in transplant tolerance. Interleukin-4 does not maintain alloantigen specific CD4(+)CD25(+) Treg. Transpl Immunol. 2013;29(1-4):51–9. doi:.https://doi.org/10.1016/j.trim.2013.10.003
  18. Liu B, Zhang H, Li J, Lu C, Chen G, Zhang G, et al. Triptolide downregulates Treg cells and the level of IL-10, TGF-β, and VEGF in melanoma-bearing mice. Planta Med. 2013;79(15):1401–7. doi:.https://doi.org/10.1055/s-0033-1350708
  19. Lasfar A, Gogas H, Zloza A, Kaufman HL, Kirkwood JM. IFN-λ cancer immunotherapy: new kid on the block. Immunotherapy. 2016;8(8):877–88. doi:.https://doi.org/10.2217/imt-2015-0021
  20. Leiva-Revilla J, Guerra-Castañon F, Olcese-Mori P, Lozada I, Rubio J, Gonzales C, et al. Efecto de la maca roja (Lepidium meyenii) sobre los niveles de IFN-γ en ratas ovariectomizadas [Effect of red maca (Lepidium meyenii) on INF-γ levels in ovariectomized rats]. Rev Peru Med Exp Salud Publica. 2014;31(4):683–8. doi:.https://doi.org/10.17843/rpmesp.2014.314.118
  21. Stern C, Kasnitz N, Kocijancic D, Trittel S, Riese P, Guzman CA, et al. Induction of CD4(+) and CD8(+) anti-tumor effector T cell responses by bacteria mediated tumor therapy. Int J Cancer. 2015;137(8):2019–28. doi:.https://doi.org/10.1002/ijc.29567
  22. Cui W, Joshi NS, Jiang A, Kaech SM. Effects of Signal 3 during CD8 T cell priming: Bystander production of IL-12 enhances effector T cell expansion but promotes terminal differentiation. Vaccine. 2009;27(15):2177–87. doi:.https://doi.org/10.1016/j.vaccine.2009.01.088
  23. Kang S, Xie J, Miao J, Li R, Liao W, Luo R. A knockdown of Maml1 that results in melanoma cell senescence promotes an innate and adaptive immune response. Cancer Immunol Immunother. 2013;62(1):183–90. doi:.https://doi.org/10.1007/s00262-012-1318-1
  24. Kiyota T, Takahashi Y, Watcharanurak K, Nishikawa M, Ohara S, Ando M, et al. Enhancement of anticancer effect of interferon-γ gene transfer against interferon-γ-resistant tumor by depletion of tumor-associated macrophages. Mol Pharm. 2014;11(5):1542–9. doi:.https://doi.org/10.1021/mp4007216
  25. Li H, Gupta S, Du WW, Yang BB. MicroRNA-17 inhibits tumor growth by stimulating T-cell mediated host immune response. Oncoscience. 2014;1(7):531–9. doi:.https://doi.org/10.18632/oncoscience.69
  26. Hsueh C, Lin JD, Wu IC, Chao TC, Yu JS, Liou MJ, et al. Vascular endothelial growth factors and angiopoietins in presentations and prognosis of papillary thyroid carcinoma. J Surg Oncol. 2011;103(5):395–9. doi:.https://doi.org/10.1002/jso.21844
  27. Costache MI, Ioana M, Iordache S, Ene D, Costache CA, Săftoiu A. VEGF Expression in Pancreatic Cancer and Other Malignancies: A Review of the Literature. Rom J Intern Med. 2015;53(3):199–208. doi:.https://doi.org/10.1515/rjim-2015-0027