Oral Exposure to Mercury Alters T Lymphocyte Phenotypes and Augments LPS-induced Cytokine Expressions in Spleen and Thymus

비장과 흉선의 림프세포와 LPS에 의해 유도된 사이토카인의 발현에 대한 수은의 영향

  • Published : 2004.08.01

Abstract

Mercury is a widespread metal and consequently there are large populations that currently exposed to low levels of mercury. Endotoxin is a component of the gram-negative bacteria and promotes inflammatory responses. The present study was designed to determine the impact of mercury on lymphocytes phenotype populations and endotoxin-induced inflammatory cytokine expressions in immune organ, spleen and thymus. Male BALB/c mice were exposed continuously to 0, 0.3, 1.5, 7.5, or 37.5 ppm of mercuric chloride in drinking water for 14 days and at the end of the treatment period, lipopolysaccharide (LPS, 0.5 mg/kg) was injected intraperitoneally 2 h prior to euthanasia. The dose-range of mercury used did not cause hepatotoxicity. Mercury at 7.5 and 37.5 ppm dose-dependently decreased CD3$^{+}$ T lymphocytes in spleen; both CD4$^{+}$ and CD8$^{+}$ single positive lymphocyte populations were decreased. Exposure to 7.5 and 37.5 ppm of mercury decreased the CD8$^{+}$ T lymphocyte population in the thymus, whereas double positive CD4$^{+}$ / CD8$^{+}$ and CD4$^{+}$ thymocytes were not altered. Mercury altered LPS-induced inflammatory cytokine gene expressions such as, tumor necrosis factor $\alpha$, interferon ${\gamma}$, and interleukin-12 in spleen and thymus. Results indicated that decreases in T lymphocyte populations in immune organs and altered cytokine gene expression may contribute to the immune-modulative effects of inorganic mercury.ganic mercury.

Keywords

References

  1. Christensen, M. M., Ellermann-Eriksen, S., Rungby, J. and Mogensen, S. C. : Influence of mercuric chloride on resistance to generalized infection with herpes simplex virus type 2 in mice. Toxicol. 114, 57 (1996) https://doi.org/10.1016/S0300-483X(96)03409-9
  2. Gerstner H. B. and Huff, J. E. : Clinical toxicology of mercury. J. Toxicol. Environ. Health 2, 491 (1977)
  3. Wild, L. G., Ortega, H. G., Lopez, M. and Salvaggio, J. E. : Immune system alteration in the rat after indirect exposure to methyl mercury chloride or methyl mercury sulfide. Environ. Res. 74, 34 (1997) https://doi.org/10.1006/enrs.1997.3748
  4. Perlingeiro, R. C. and Queiroz, M. L. : Polymorphonuclear phagocytosis and killing in workers exposed to inorganic mercury. Int. J. Immunopharmacol. 16, 1011 (1994) https://doi.org/10.1016/0192-0561(94)90080-9
  5. Koller, L. D. : Methylmercury: effect on oncogenic and nononcogenic viruses in mice. Am. J. Vet. Res.36, 1501 (1975)
  6. Sapin, C., Druet, E. and Druet, P. : Induction of anti-glomerular basement membrane antibodies in the Brown-Norway rat by mercuric chloride. Clin. Exp. Immunol . 28, 173 (1977)
  7. Kim, S. H., Johnson, V. J. and Sharma, R. P. : Mercury inhibits nitric oxide production but activates proinflammatory cytokine expression in murine macrophage: differential modulation of NF-kappaB and p38 MAPK signaling pathways. Nitric Oxide 7, 67 (2002) https://doi.org/10.1016/S1089-8603(02)00008-3
  8. Hu, H., Moller, G. and Abedi-Valugerdi, M. : Major histocompatibility complex class II antigens are required for both cytokine production and proliferation induced by mercuric chloride in vitro. J. Autoimmun . 10, 441 (1997) https://doi.org/10.1006/jaut.1997.9997
  9. Nielsen, J. B. and Andersen, O. : Oral mercuric chloride exposure in mice: effects of dose on intestinal absorbtion and relative organ distribution. Toxicology 59, 1 (1989) https://doi.org/10.1016/0300-483X(89)90152-2
  10. Johnson, V. J. and Sharma, R. P. : Gender-dependent immunosuppression following subacute exposure to fumonisin B1. Int. lmmunpharmacol . 1, 2023 (2001)
  11. Tsunoda, M. and Sharma, R. P. : Modulation of tumor necrosis factor alpha expression in mouse brain after exposure to aluminum in drinking water. Arch. Toxicol. 73, 419 (1999) https://doi.org/10.1007/s002040050630
  12. Kosuda, L. L., Whalen, B., Greiner, D. L. and Bigazzi, P. E. : Mercury-induced autoimmunity in Brown Norway rats: kinetics of changes in RT6+ T lymphocytes correlated with IgG isotypes of circulating autoantibodies to laminin 1. Toxicol. 125, 215 (1998)
  13. Koller, L. D. : Immunotoxicology of heavy metals. J.Int. J. Immunpharmacol. 2, 269 (1980) https://doi.org/10.1016/0192-0561(80)90027-2
  14. National Toxicology Program: Toxicology and carcinogenesis studies of mercuric chloride in F344/N rats and B6CF1 mice. NlH publication, p. 91 (1993)
  15. Shen, X., Lee, K. and Konig, R. : Effects of heavy metal ions on resting and antigen-activated CD4(+) T cells. Toxicol. 169, 67 (2001) https://doi.org/10.1016/S0300-483X(01)00483-8
  16. Ghosh, S., Latimer, R. D., Gray, B. M., Harwood, R. J. and Oduro, A. : Endotoxin-induced organ injury. Crit. Care Med. 21, S19 (1993)
  17. Spitzer J. A., Zhang, P. and Mayer; A. M. : Functional characterization of peripheral circulating and liver recruited neutrophils in endotoxic rats. J. Leukoc. Biol. 56, 166 (1994)
  18. Tsutsui, H., Matsui, K., Kawada, N., Hyodo, Y., Hayashi, N., Okamura, H., Higashino, K. and Akanishi, K. : IL-18 accounts for both TNF-alpha and Fas ligand-mediated hepatotoxic pathways in endotoxin-induced liver injury in mice. J. Immunol. 159, 3961 (1997)
  19. Trinchieri, G. : Interleukin-12 and the regulation of innate resistance and adaptive immunity. Nat. Rev. Immunol. 3(2), 133 (2003)
  20. Godfrey, D. I., Kennedy, J., Gately, M. K., Hakimi , J., Hubbard, B. R. and Zlotnik, A. : IL-12 influences intrathymic T cell development. J. Immunol. 152, 2729 (1994)
  21. Bigazzi, P. E. : Mercury. In: Zelikoff, J. and Thomas, P. (Eds.), Metal Immunotoxicology. Taylor and Francis, london, UK. p.131 (1998)