7 research outputs found

    Inflammatory Biomarkers of Hydrogen Sulfide Induced Neurotoxicity and Degeneration

    Get PDF
    Hydrogen sulfide (H2S) has dual actions in the human body as an endogenously produced signaling molecule and a toxic xenobiotic. It is a byproduct of several industries, including oil, petroleum and paper milling and natural forms of exposure are of concern for individuals living near landfills or volcanos. That hydrogen sulfide has potential for use as a chemical weapon is disconcerting. The gas is colorless and has a characteristic rotten egg odor. Acute H2S intoxication can lead to both short and long term neurological sequelae, including neurodegeneration, memory and motor impairment, however, the underlying mechanisms are still unknown. Our hypothesis is that neuroinflammation, characterized by an intense inflammatory response from activated glial cells, can cause cell death by invoking the production of many pro and anti-inflammatory cytokines. To test this hypothesis we used histopathology to visualize the affected tissue; then measured cytokines in the brain tissue and serum of mice exposed to H2S by inhalation. Results show increased glial fibrillary protein levels, indicating a recruitment of reactive astrocytes to the tissue, starting around day 3 post exposure. Understanding the basic mechanisms underlying neuroinflammation contributes to our long term objective of discovering countermeasures against and treatment of H2S induced neurodegeneration

    Midazolam Efficacy Against Acute Hydrogen Sulfide-Induced Mortality and Neurotoxicity.

    Get PDF
    Hydrogen sulfide (H2S) is a colorless, highly neurotoxic gas. It is not only an occupational and environmental hazard but also of concern to the Department of Homeland Security for potential nefarious use. Acute high-dose H2S exposure causes death, while survivors may develop neurological sequelae. Currently, there is no suitable antidote for treatment of acute H2S-induced neurotoxicity. Midazolam (MDZ), an anti-convulsant drug recommended for treatment of nerve agent intoxications, could also be of value in treating acute H2S intoxication. In this study, we tested the hypothesis that MDZ is effective in preventing/treating acute H2S-induced neurotoxicity. This proof-of-concept study had two objectives: to determine whether MDZ prevents/reduces H2S-induced mortality and to test whether MDZ prevents H2S-induced neurological sequelae. MDZ (4 mg/kg) was administered IM in mice, 5 min pre-exposure to a high concentration of H2S at 1000 ppm or 12 min post-exposure to 1000 ppm H2S followed by 30 min of continuous exposure. A separate experiment tested whether MDZ pre-treatment prevented neurological sequelae. Endpoints monitored included assessment of clinical signs, mortality, behavioral changes, and brain histopathological changes. MDZ significantly reduced H2S-induced lethality, seizures, knockdown, and behavioral deficits (p < 0.01). MDZ also significantly prevented H2S-induced neurological sequelae, including weight loss, behavior deficits, neuroinflammation, and histopathologic lesions (p < 0.01). Overall, our findings show that MDZ is a promising drug for reducing H2S-induced acute mortality, neurotoxicity, and neurological sequelae

    Inflammatory Biomarkers of Hydrogen Sulfide Induced Neurotoxicity and Degeneration

    No full text
    Hydrogen sulfide (H2S) has dual actions in the human body as an endogenously produced signaling molecule and a toxic xenobiotic. It is a byproduct of several industries, including oil, petroleum and paper milling and natural forms of exposure are of concern for individuals living near landfills or volcanos. That hydrogen sulfide has potential for use as a chemical weapon is disconcerting. The gas is colorless and has a characteristic rotten egg odor. Acute H2S intoxication can lead to both short and long term neurological sequelae, including neurodegeneration, memory and motor impairment, however, the underlying mechanisms are still unknown. Our hypothesis is that neuroinflammation, characterized by an intense inflammatory response from activated glial cells, can cause cell death by invoking the production of many pro and anti-inflammatory cytokines. To test this hypothesis we used histopathology to visualize the affected tissue; then measured cytokines in the brain tissue and serum of mice exposed to H2S by inhalation. Results show increased glial fibrillary protein levels, indicating a recruitment of reactive astrocytes to the tissue, starting around day 3 post exposure. Understanding the basic mechanisms underlying neuroinflammation contributes to our long term objective of discovering countermeasures against and treatment of H2S induced neurodegeneration.</p

    Characterizing a mouse model for evaluation of countermeasures against hydrogen sulfide–induced neurotoxicity and neurological sequelae

    No full text
    Hydrogen sulfide (H2 S) is a highly neurotoxic gas. It is the second most common cause of gas-induced deaths. Beyond mortality, surviving victims of acute exposure may suffer long-term neurological sequelae. There is a need to develop countermeasures against H2 S poisoning. However, no translational animal model of H2 S-induced neurological sequelae exists. Here, we describe a novel mouse model of H2 S-induced neurotoxicity for translational research. In paradigm I, C57/BL6 mice were exposed to 765 ppm H2 S for 40 min on day 1, followed by 15-min daily exposures for periods ranging from 1 to 6 days. In paradigm II, mice were exposed once to 1000 ppm H2 S for 60 minutes. Mice were assessed for behavioral, neurochemical, biochemical, and histopathological changes. H2 S intoxication caused seizures, dyspnea, respiratory depression, knockdowns, and death. H2 S-exposed mice showed significant impairment in locomotor and coordinated motor movement activity compared with controls. Histopathology revealed neurodegenerative lesions in the collicular, thalamic, and cortical brain regions. H2 S significantly increased dopamine and serotonin concentration in several brain regions and caused time-dependent decreases in GABA and glutamate concentrations. Furthermore, H2 S significantly suppressed cytochrome c oxidase activity and caused significant loss in body weight. Overall, male mice were more sensitive than females. This novel translational mouse model of H2 S-induced neurotoxicity is reliable, reproducible, and recapitulates acute H2 S poisoning in humans
    corecore