Immunotoxicology
Effect of 2,3,7,8-tetrachlorodibenzo-p-dioxin on anti-CD3-induced changes in T-cell subsets and cytokine production

https://doi.org/10.1016/0192-0561(95)00080-1Get rights and content

Abstract

The influence of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) exposure on the cytokine-dependent toxicity syndrome induced by the injection of 145-2C11 (anti-CD3), a hamster monoclonal antibody to the CD3 epsilon portion of the murine T-cell receptor, was studied. This syndrome has been attributed to the transient release of several cytokines including TNF-α, IFN-γ, IL-2, IL-3, IL-6, and GM-CSF. Exposure of C57B1/6 mice to TCDD (15 μg/kg) 2 days prior to anti-CD3 injection exacerbated anti-CD3-induced toxicity as evidenced by significantly enhanced and prolonged body weight loss and lymphoid tissue atrophy. Unexpectedly, TCDD exposure did not alter plasma levels of TNF or IL-2 at any time after anti-CD3 injection. However, plasma IFN-γ was significantly reduced at 24 h and plasma IL-6 levels were elevated 48 h after anti-CD3 injection in TCDD-treated mice. In addition, TCDD exposure resulted in elevated levels of plasma GM-CSF at 24 and 48 h. Since the body weight of TCDD-treated mice diverged from vehicle-treated mice at 48 h, it suggests that the increased IL-6 and GM-CSF may have contributed to the prolonged loss of body weight. The ability of spleen cells from vehicle- and TCDD-treated mice to produce cytokines was evaluated in vitro at various times after anti-CD3 injection. TCDD treatment resulted in reduced IL-2 and GM-CSF production at 90 min but increased GM-CSF production at 48 h post-anti-CD3 injection. In contrast, TCDD exposure did not influence cytokine production by spleen cells from mice injected with a control IgG and activated in vitro with anti-CD3. Flow cytometric analysis showed that the percentage of CD4+ cells in the draining lymph nodes from TCDD-treated mice was reduced 48–144 h post-anti-CD3 injection. In contrast, the percentage of CD8+ cells was not affected by TCDD exposure. A high fraction of lymph node cells (LNC) from TCDD-treated animals showed decreased forward angle light scatter and increased 90° light scatter following anti-CD3 injection, which is a pattern characteristic of cells undergoing apoptosis. In contrast, few LNC from vehicle-treated animals showed this light scatter profile. These data suggest that TCDD may be targeting T-helper cells during activation resulting in activation-driven cell death (apoptosis) rather than differentiation.

References (40)

  • W. Swat et al.

    Detection of apoptosis of immature CD48+ thymocytes by flow cytometry

    J. immun. Meth.

    (1991)
  • M.J. Taylor et al.

    Inhibition of acute TCDD toxicity by treatment with anti-tumor necrosis antibody or dexamethasone

    Toxic. appl. Pharmac.

    (1992)
  • R.S. Tomar et al.

    Reduced T-helper cell function in mice exposed to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)

    Toxic. Lett.

    (1991)
  • J.G. Vos et al.

    Immune alterations

  • M.-L. Alegre et al.

    Cytokine release syndrome induced by the 145-2C11 anti-CD3 monoclonal antibody in mice: prevention by high doses of methylprednisolone

    J. Immun.

    (1991)
  • M. Alegre et al.

    Hypothermia and hypoglycemia induced by anti-CD3 monoclonal antibody in mice: role of tumor necrosis factor

    Eur. J. Immun.

    (1990)
  • D.W. Bombick et al.

    2,3,7,8-Tetrachlorodibenzo-p-dioxin causes elevation of the levels of the protein tyrosine kinase pp60c-src

    J. Biochem. Toxic.

    (1987)
  • D.A. Clark et al.

    Enhanced suppressor cell activity as a mechanism of immunosuppression by 2,3,7,8-tetrachlorodibenzo-p-dioxin

  • G.C. Clark et al.

    Tumor necrosis factor involvement in the toxicity of TCDD: the role of endotoxin in the response

    Exp. clin. Immunogenet.

    (1994)
  • C. Ferran et al.

    Anti-tumor necrosis factor modulates anti-CD3-triggered T-cell cytokine gene expression in vivo

    J. clin. Invest.

    (1994)
  • Cited by (47)

    • Immunotoxicology of Halogenated Aromatic Hydrocarbons

      2018, Comprehensive Toxicology: Third Edition
    • AHR-mediated immunomodulation: The role of altered gene transcription

      2009, Biochemical Pharmacology
      Citation Excerpt :

      For example, interferon gamma (IFNγ) production in response to influenza virus infection is suppressed in the lymph nodes but augmented 10-fold in the lungs of TCDD-treated mice [7,10]. Similarly, TNFα production is not altered in anti-CD3 treated mice [11], whereas it is decreased in P815 tumor-injected mice [12] and significantly increased in LPS-treated mice [13]. These differential effects of TCDD on a single cytokine reflect the different types of cells that are capable of making the cytokine, the state of activation of the cells, and the nature of the activation stimulus.

    • Fewer CTL, not enhanced NK cells, are sufficient for viral clearance from the lungs of immunocompromised mice

      2003, Cellular Immunology
      Citation Excerpt :

      Exposure to TCDD has been shown to alter the expression of genes encoding anti-viral cytokines [45,46,48,53,54]. In fact, the idea that activation of the AhR dysregulates cytokine production and that these defects underlie the immunomodulatory activity of AhR ligands is supported by a large number of studies [23,55–58]. Therefore, a cytokine-mediated compensatory mechanism was logical.

    View all citing articles on Scopus
    View full text