Prenatal ethanol exposure is the leading known cause of mental retardation. Growing evidence suggests that excessive cell death is a major component of the pathogenesis of ethanol-induced birth defects. However, there is a fundamental gap in understanding how ethanol leads to apoptotic cell death in embryos. Continued existence of this gap represents an important problem because, until it is filled, understanding of ethanol- induced apoptosis that leads to teratogenesis will remain largely incomprehensible. Our long-term goal is directed toward the development of effective strategies against ethanol's teratogenesis; strategies based on prevention of ethanol-induced apoptosis through targeting specific proteins involved in apoptosis. The overall objective of this particular proposal is to establish Siah1, a member of the seven in absentia homology family, as a feasible target for the prevention of ethanol-induced apoptosis and teratogenesis. The central hypothesis to be tested is that Siah1 mediates ethanol-induced apoptosis in neural crest cells (NCCs) by activating the p53 pathway and that the inhibition of Siah1-mediated apoptosis can prevent ethanol-induced teratogenesis. Our hypothesis has been formulated on the basis of strong preliminary data produced in our laboratory. To test our hypothesis, the following specific aims will be addressed:
Aim1 : To characterize the role of Siah1 in ethanol-induced apoptosis in NCCs. We will determine the effects of ethanol on Siah1 mRNA and protein expression, and investigate the potential of ethanol to promote Siah1 nuclear translocation and induce apoptosis.
Aim2 : To test the hypothesis that Siah1 mediates ethanol-induced apoptosis in NCCs by activating the p53 pathway. This will be accomplished by determining the role of Siah1 in ethanol-induced activation of p53 signaling pathway and in the induction of p53 downstream proapoptotic proteins and apoptosis.
Aim3 : To define the role of the inhibition of Siah1 function in conferring protection against ethanol-induced teratogenesis. This will be accomplished by using whole embryo culture, Siah1 knockout mice and an oral intake FASD model. We will determine whether knocking down Siah1 diminishes ethanol-induced malformations in cultured mouse embryos and whether Siah1 deficiency in Siah1-/- mouse embryos can confer in vivo protection against ethanol-induced teratogenesis. The proposed work is innovative, because it focuses on a novel approach, targeting specific proteins directly involved in the apoptotic pathway, to preventing ethanol-induced apoptosis and teratogenesis. The theoretical concept described in the application is also highly innovative because this is the first study attempting to prevent ethanol-induced apoptosis and teratogenesis specifically through the newly recognized actions of Siah1 in apoptosis. The results from this study will be significant, because the insights gained by the accomplishment of these aims will help in elucidating the role of Siah1 in mediating ethanol- induced teratogenesis. They are also expected to yield strategies for the prevention of ethanol's teratogenesis and to fundamentally advance the field of FASD research.

Public Health Relevance

The proposed research is relevant to public health because the insights gained by the accomplishments of the proposed studies will help in elucidating the mechanisms underlying ethanol-induced cell death, and validating possible molecular targets for the development of novel therapeutics for the prevention of human Fetal Alcohol Spectrum Disorders (FASD). Thus, the proposed research is relevant to the NIAAA's mission that pertains to using knowledge gained in uncovering target sites for alcohol's action on the embryonic and fetal stages of life to begin developing potential therapeutic or preventative interventions.

Agency
National Institute of Health (NIH)
Institute
National Institute on Alcohol Abuse and Alcoholism (NIAAA)
Type
Research Project (R01)
Project #
5R01AA020265-05
Application #
9108234
Study Section
Neurotoxicology and Alcohol Study Section (NAL)
Program Officer
Hereld, Dale
Project Start
2012-07-05
Project End
2017-06-30
Budget Start
2016-07-01
Budget End
2017-06-30
Support Year
5
Fiscal Year
2016
Total Cost
Indirect Cost
Name
University of Louisville
Department
Pharmacology
Type
Schools of Medicine
DUNS #
057588857
City
Louisville
State
KY
Country
United States
Zip Code
40208
Yuan, Fuqiang; Chen, Xiaopan; Liu, Jie et al. (2018) Sulforaphane restores acetyl-histone H3 binding to Bcl-2 promoter and prevents apoptosis in ethanol-exposed neural crest cells and mouse embryos. Exp Neurol 300:60-66
Wang, Keling; Chen, Xiaopan; Liu, Jie et al. (2018) Embryonic exposure to ethanol increases the susceptibility of larval zebrafish to chemically induced seizures. Sci Rep 8:1845
Yuan, Fuqiang; Chen, Shao-Yu (2018) Manipulation of MicroRNAs in Cultured Mouse Embryos: Applications for Developmental Toxicology. Methods Mol Biol 1797:205-214
Dou, Xiaowei; Menkari, Carrie; Mitsuyama, Rei et al. (2018) L1 coupling to ankyrin and the spectrin-actin cytoskeleton modulates ethanol inhibition of L1 adhesion and ethanol teratogenesis. FASEB J 32:1364-1374
Barve, Shirish; Chen, Shao-Yu; Kirpich, Irina et al. (2017) Development, Prevention, and Treatment of Alcohol-Induced Organ Injury: The Role of Nutrition. Alcohol Res 38:289-302
Lee, Philbert; Jiang, Shangwen; Li, Yuanyuan et al. (2017) Phosphorylation of Pkp1 by RIPK4 regulates epidermal differentiation and skin tumorigenesis. EMBO J 36:1963-1980
Ma, Yanlei; Yue, Jiping; Zhang, Yao et al. (2017) ACF7 regulates inflammatory colitis and intestinal wound response by orchestrating tight junction dynamics. Nat Commun 8:15375
Yuan, Fuqiang; Chen, Xiaopan; Liu, Jie et al. (2017) Up-regulation of Siah1 by ethanol triggers apoptosis in neural crest cells through p38 MAPK-mediated activation of p53 signaling pathway. Arch Toxicol 91:775-784
Xu, Zheng; Tong, Qian; Zhang, Zhiguo et al. (2017) Inhibition of HDAC3 prevents diabetic cardiomyopathy in OVE26 mice via epigenetic regulation of DUSP5-ERK1/2 pathway. Clin Sci (Lond) 131:1841-1857
Yue, Jiping; Zhang, Yao; Liang, Wenguang G et al. (2016) In vivo epidermal migration requires focal adhesion targeting of ACF7. Nat Commun 7:11692

Showing the most recent 10 out of 17 publications