Abstract

BackgroundThe role of sex hormones on cellular function is unclear. Studies show androgens and estrogens are protective in the CNS, whereas other studies found no effects or damaging effects. Furthermore, sex differences have been observed in multiple oxidative stress-associated CNS disorders, such as Alzheimer’s disease, depression, and Parkinson’s disease. The goal of this study is to examine the relationship between sex hormones (i.e., androgens and estrogens) and oxidative stress on cell viability.MethodsN27 and PC12 neuronal and C6 glial phenotypic cell lines were used. N27 cells are female rat derived, whereas PC12 cells and C6 cells are male rat derived. These cells express estrogen receptors and the membrane-associated androgen receptor variant, AR45, but not the full-length androgen receptor. N27, PC12, and C6 cells were exposed to sex hormones either before or after an oxidative stressor to examine neuroprotective and neurotoxic properties, respectively. Estrogen receptor and androgen receptor inhibitors were used to determine the mechanisms mediating hormone-oxidative stress interactions on cell viability. Since the presence of AR45 in the human brain tissue was unknown, we examined the postmortem brain tissue from men and women for AR45 protein expression.ResultsNeither androgens nor estrogens were protective against subsequent oxidative stress insults in glial cells. However, these hormones exhibited neuroprotective properties in neuronal N27 and PC12 cells via the estrogen receptor. Interestingly, a window of opportunity exists for sex hormone neuroprotection, wherein temporary hormone deprivation blocked neuroprotection by sex hormones. However, if sex hormones are applied following an oxidative stressor, they exacerbated oxidative stress-induced cell loss in neuronal and glial cells.ConclusionsSex hormone action on cell viability is dependent on the cellular environment. In healthy neuronal cells, sex hormones are protective against oxidative stress insults via the estrogen receptor, regardless of sex chromosome complement (XX, XY). However, in unhealthy (e.g., high oxidative stress) cells, sex hormones exacerbated oxidative stress-induced cell loss, regardless of cell type or sex chromosome complement. The non-genomic AR45 receptor, which is present in humans, mediated androgen’s damaging effects, but it is unknown which receptor mediated estrogen’s damaging effects. These differential effects of sex hormones that are dependent on the cellular environment, receptor profile, and cell type may mediate the observed sex differences in oxidative stress-associated CNS disorders.

Highlights

  • The role of sex hormones on cellular function is unclear

  • Sex hormones are protective against oxidative stress insults via the estrogen receptor, regardless of sex chromosome complement (XX, XY)

  • The nongenomic AR45 receptor, which is present in humans, mediated androgen’s damaging effects, but it is unknown which receptor mediated estrogen’s damaging effects

Read more

Summary

Introduction

The role of sex hormones on cellular function is unclear. Studies show androgens and estrogens are protective in the CNS, whereas other studies found no effects or damaging effects. Sex differences have been observed in multiple oxidative stress-associated CNS disorders, such as Alzheimer’s disease, depression, and Parkinson’s disease. The goal of this study is to examine the relationship between sex hormones (i.e., androgens and estrogens) and oxidative stress on cell viability. Alzheimer’s disease [6], major depression [7, 8], and stress disorders [9, 10] are more prevalent in women than men. Menopause in women influences the prevalence of CNS disorders, such as Alzheimer’s disease [11], stroke [12], Parkinson’s disease [13,14,15], depression [16], anxiety disorders [17], and schizophrenia [18, 19]. Testosterone levels (8–60 ng/ dL) are maintained in women during menopause [21,22,23]; these testosterone levels are 27–30 fold less than the levels observed in healthy young adult men (240–950 ng/dL) and middle-aged men (219–929 ng/dL) [24, 25]

Objectives
Methods
Results
Discussion
Conclusion
Full Text
Paper version not known

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call