ALS, commonly known as Lou Gehrig's disease, is the most common adult motor neuron disease. The disease's primary hallmark is the selective dysfunction and death of the neurons in the motor pathways. Although approximately 10% of ALS cases are inherited (familial), the majority of cases have no genetic component identified (sporadic) and exposure to yet unidentified environmental toxicants might be responsible for these cases. Among the familial cases, approximately 20% are caused by dominantly inherited mutations in the Cu, Zn superoxide dismutase (SOD1). Rodents overexpressing human mutant SOD1 generally develop an ALS-like phenotype. Several hypotheses, including oxidative stress induced by SOD1 aberrant catalysis, and mitochondrial dysfunction have been proposed to explain the toxic effect of mutant SOD1. To explore the role of antioxidant defenses in ALS, knockout mice were employed using the glutamate-cysteine ligase modifier subunit (GCLM-/-), which have a 70-80% reduction of total glutathione (GSH) when compared to wild-type littermates. Although GCLM-/- mice are fertile and viable, the life span of GCLM(-/-)/hSOD1G93A mice decreased in 50-60% when compared to the GCLM(+/+)/hSOD1G93A mice. The decrease in life span seems to be associated with aggravated mitochondrial pathology commonly observed in transgenic mice overexpressing mutated forms of hSOD1 that retain superoxide dismutase activity. Interestingly, when the GCLM-/- animals were mated with a different ALS animal model which overexpresses a mutant hSOD1 with undetectable dismutase activity (hSOD1H46R/H48Q), no effect was observed in the onset or survival of GCLM(-/-)/hSOD1H46R/H48Q mice. In addition, little or no mitochondrial pathology was observed in these animals. On the aforementioned context, the specific aims of the proposal are: 1: To evaluate the effect of reduced GSH levels on the onset and progression of the disease in hSOD1G93A, hSOD1H46R/H48Q and hSOD1WTmice;2: To determine the role of hSOD1-induced mitochondrial dysfunction in the toxicity of astrocytes expressing ALS-linked mutant SOD1s toward co-cultured motor neurons;and 3. To evaluate the effect of increased mitochondrial peroxide detoxification in the onset and progression of disease in hSOD1G93A and hSOD1H46R/H48Q mice. The proposal is designed to determine the effect of reduced GSH content in mitochondrial pathology and disease progression in ALS. Because a specific modifier, such as GSH deficiency, may affect only certain SOD1 mutants, the result of these experiments will contribute to understand the potential difference in the molecular pathways by which different SOD1 mutants produce disease. Since surrounding glial cells also play a key role in the disease progression, the results obtained should also identify cell-type specific effects of GSH deficiency and hSOD1 toxicity that will be critical to the understanding of ALS pathogenesis. Finally, the results obtained from two different strategies designed to increase mitochondrial peroxide detoxification capacity will help delineate the value of mitochondrial-targeted antioxidants as a viable clinical therapy for ALS.

Public Health Relevance

Amyotrophic lateral sclerosis (ALS) or Lou Gehrig's disease is characterized by the specific death of the nerve cells that control muscle movement, leading to paralysis and eventual death. Malfunction of the mitochondria, the powerhouse of the cell, may be responsible for the observed neurodegeneration. This proposal will investigate the role of glutathione, a natural antioxidant found in the cells, as a major protective factor for mitochondrial function. One will also investigate possible ways to restore normal mitochondrial function as a therapy for ALS.

Agency
National Institute of Health (NIH)
Institute
National Institute of Environmental Health Sciences (NIEHS)
Type
Career Transition Award (K99)
Project #
1K99ES019186-01
Application #
7962876
Study Section
Special Emphasis Panel (ZES1-LWJ-V (01))
Program Officer
Shreffler, Carol K
Project Start
2010-12-13
Project End
2012-07-31
Budget Start
2010-12-13
Budget End
2012-07-31
Support Year
1
Fiscal Year
2011
Total Cost
$90,000
Indirect Cost
Name
University of Wisconsin Madison
Department
Pharmacology
Type
Schools of Pharmacy
DUNS #
161202122
City
Madison
State
WI
Country
United States
Zip Code
53715
Killoy, Kelby M; Harlan, Benjamin A; Pehar, Mariana et al. (2018) Decreased glutathione levels cause overt motor neuron degeneration in hSOD1WT over-expressing mice. Exp Neurol 302:129-135
Diaz-Amarilla, Pablo; Miquel, Ernesto; Trostchansky, Andrés et al. (2016) Electrophilic nitro-fatty acids prevent astrocyte-mediated toxicity to motor neurons in a cell model of familial amyotrophic lateral sclerosis via nuclear factor erythroid 2-related factor activation. Free Radic Biol Med 95:112-20
Pehar, Mariana; Ball, Lauren E; Sharma, Deep R et al. (2016) Changes in Protein Expression and Lysine Acetylation Induced by Decreased Glutathione Levels in Astrocytes. Mol Cell Proteomics 15:493-505
Pehar, Mariana; Beeson, Gyda; Beeson, Craig C et al. (2014) Mitochondria-targeted catalase reverts the neurotoxicity of hSOD1G?³A astrocytes without extending the survival of ALS-linked mutant hSOD1 mice. PLoS One 9:e103438
Vargas, Marcelo R; Johnson, Delinda A; Johnson, Jeffrey A (2011) Decreased glutathione accelerates neurological deficit and mitochondrial pathology in familial ALS-linked hSOD1(G93A) mice model. Neurobiol Dis 43:543-51