Mercury (Hg) toxicity affects numerous organ systems including the immune system. The immunotoxicity of Hg is complex, in that, immunosuppression and immunostimulation occur upon exposure to this metal, and both of these outcomes may have adverse effects on human health. This proposal is constructed around the conceptual framework that Hg is an environmental agent that contributes to the development of autoimmune disease. Animal studies have established a connection between experimental exposure to Hg and lupus-like autoimmune disease. Similarly, case reports demonstrating a correlation between accidental Hg exposure and either the onset or the severity of autoimmune disease symptoms support a link between Hg intoxication and the etiology of human autoimmune disease. From this perspective, Hg is one of the few suspected environmental agents where a link between exposure to the agent and autoimmune disease has been indicated. However, the mechanisms whereby Hg initiates, potentiates, or perpetuates autoimmune responses have not been established. In particular, there is a dearth of information regarding the biochemical/molecular mechanisms involved in Hg-mediated autoimmunity. Accordingly, the long-term goal of our research is to identify biochemical/molecular mechanisms underlying Hg-mediated autoimmune disease. Non-cytotoxic concentrations of Hg2+ interfere with the CD95 apoptotic signaling pathway and also interfere with protein tyrosine mediated signaling. Thus, this proposal focuses on lymphocyte signaling pathways involved in the regulation of CD95-induced apoptosis. Since CD95-mediated apoptosis is involved in the regulation of peripheral tolerance, dysregulation of CD95-signaling by Hg may be one factor (together with other genetic or environmental factors) that contributes to autoimmune disease. The hypothesis guiding this proposal is that Hg2+ disrupts CD95-mediated apoptosis, which contributes to a breakdown in peripheral tolerance to autoantigens leading to the development of autoimmune disease. Using cell lines and human CD4+ lymphoblasts this hypothesis will be tested by determining; 1) the influence of Hg2+ on components of the CD95 apoptotic machinery and 2) the effects of Hg2+ on non-receptor kinase signaling pathways that may interact with and regulate CD95. A third specific aim will employ a mouse model to test whether disruption of CD95-mediated cell death by Hg intoxication in vivo enhances the survival of autoreactive T lymphocytes.

Agency
National Institute of Health (NIH)
Institute
National Institute of Environmental Health Sciences (NIEHS)
Type
Research Project (R01)
Project #
5R01ES012403-04
Application #
7017805
Study Section
Alcohol and Toxicology Subcommittee 4 (ALTX)
Program Officer
Mastin, Patrick
Project Start
2003-04-03
Project End
2008-01-31
Budget Start
2006-02-01
Budget End
2008-01-31
Support Year
4
Fiscal Year
2006
Total Cost
$255,367
Indirect Cost
Name
University of Rochester
Department
Public Health & Prev Medicine
Type
Schools of Dentistry
DUNS #
041294109
City
Rochester
State
NY
Country
United States
Zip Code
14627
Ziemba, Stamatina E; Menard, Sherri L; McCabe Jr, Michael J et al. (2009) T-cell receptor signaling is mediated by transient Lck activity, which is inhibited by inorganic mercury. FASEB J 23:1663-71
Farrer, David G; Hueber, Sara; Laiosa, Michael D et al. (2008) Reduction of myeloid suppressor cell derived nitric oxide provides a mechanistic basis of lead enhancement of alloreactive CD4(+) T cell proliferation. Toxicol Appl Pharmacol 229:135-45
Laiosa, Michael D; Eckles, Kevin G; Langdon, Margaret et al. (2007) Exposure to inorganic mercury in vivo attenuates extrinsic apoptotic signaling in Staphylococcal aureus enterotoxin B stimulated T-cells. Toxicol Appl Pharmacol 225:238-50
McCabe Jr, Michael J; Laiosa, Michael D; Li, Li et al. (2007) Low and nontoxic inorganic mercury burdens attenuate BCR-mediated signal transduction. Toxicol Sci 99:512-21
Ziemba, Stamatina E; Mattingly, Raymond R; McCabe Jr, Michael J et al. (2006) Inorganic mercury inhibits the activation of LAT in T-cell receptor-mediated signal transduction. Toxicol Sci 89:145-53
Ziemba, Stamatina E; McCabe Jr, Michael J; Rosenspire, Allen J (2005) Inorganic mercury dissociates preassembled Fas/CD95 receptor oligomers in T lymphocytes. Toxicol Appl Pharmacol 206:334-42
McCabe Jr, Michael J; Eckles, Kevin G; Langdon, Margaret et al. (2005) Attenuation of CD95-induced apoptosis by inorganic mercury: caspase-3 is not a direct target of low levels of Hg2+. Toxicol Lett 155:161-70
McCabe Jr, Michael J; Whitekus, Michael J; Hyun, Joogyung et al. (2003) Inorganic mercury attenuates CD95-mediated apoptosis by interfering with formation of the death inducing signaling complex. Toxicol Appl Pharmacol 190:146-56