The hormonal control of cell death is currently the best established mechanism for creating sex differences in the nervous system. Nonetheless, very little is known about how hormones such as testosterone regulate neuronal cell death. This project will examine the cellular and molecular mechanisms underlying hormonally controlled cell death in three well-studied model systems: the spinal nucleus of the bulbocavernosus (SNB), the anteroventral periventricular nucleus (AVPv) of the hypothalamus, and the bed nucleus of the stria terminalis (BNST). Mice will be used throughout, to take advantage of the power of genetically manipulated strains. Members of the Bcl-2 family of proteins are crucial regulators of cell death in many neural areas.
In Aim I we will ask whether Bcl-2 family members also regulate sexually dimorphic cell death by examining the expression and hormone regulation of pro-life (Bcl-2, Bcl-xL) and pro-death (Bax) molecules in the brain and spinal cord of neonatal mice. Testosterone decreases developmental cell death of the SNB and BNST, while increasing cell death in AVPv. We therefore predict that hormone treatments will differentially affect death-regulatory proteins in these neural areas. Bax knockout mice will be used in Aims 2 and 3 to test the hypotheses that Bax is required for the death of neurons in SNB, AVPv, and BNST; if so, then sex differences in neuron number in these regions will be eliminated in Bax knockouts. In addition to sex differences in overall cell number, AVPv and BNST exhibit large sex differences in the expression of neurotransmitters or neuropeptides. It is not known whether these differences are due to cell death, or the specification of neuronal phenotype. Cell death mutant mice will be used to discriminate between these hypotheses. Finally, in the SNB, testosterone likely regulates neuronal death indirectly, by controlling the availability of neurotrophic factors from target cells.
Aim 4 will test whether a recently identified trophic factor rescues SNB cells of mice and, in so doing, alters the expression of Bcl-2 family members. Together, these studies will allow us to specify at a mechanistic level how hormones control neuronal cell death in neural systems. This work is relevant to understanding sex differences in susceptibility to human neurodevelopmental disorders and neurodegenerative diseases in adulthood.

Agency
National Institute of Health (NIH)
Institute
National Institute of Mental Health (NIMH)
Type
Research Project (R01)
Project #
5R01MH068482-04
Application #
7233123
Study Section
Neurodegeneration and Biology of Glia Study Section (NDBG)
Program Officer
Panchision, David M
Project Start
2004-04-01
Project End
2009-03-31
Budget Start
2007-04-01
Budget End
2009-03-31
Support Year
4
Fiscal Year
2007
Total Cost
$236,613
Indirect Cost
Name
University of Massachusetts Amherst
Department
Psychology
Type
Schools of Arts and Sciences
DUNS #
153926712
City
Amherst
State
MA
Country
United States
Zip Code
01003
Strahan, J Alex; Walker 2nd, William H; Montgomery, Taylor R et al. (2017) Minocycline causes widespread cell death and increases microglial labeling in the neonatal mouse brain. Dev Neurobiol 77:753-766
Mosley, Morgan; Weathington, Jill; Cortes, Laura R et al. (2017) Neonatal Inhibition of DNA Methylation Alters Cell Phenotype in Sexually Dimorphic Regions of the Mouse Brain. Endocrinology 158:1838-1848
Mosley, Morgan; Shah, Charisma; Morse, Kiriana A et al. (2017) Patterns of cell death in the perinatal mouse forebrain. J Comp Neurol 525:47-64
Forger, Nancy G; Strahan, J Alex; Castillo-Ruiz, Alexandra (2016) Cellular and molecular mechanisms of sexual differentiation in the mammalian nervous system. Front Neuroendocrinol 40:67-86
Shen, Erica Y; Ahern, Todd H; Cheung, Iris et al. (2015) Epigenetics and sex differences in the brain: A genome-wide comparison of histone-3 lysine-4 trimethylation (H3K4me3) in male and female mice. Exp Neurol 268:21-9
Ahern, Todd H; Krug, Stefanie; Carr, Audrey V et al. (2013) Cell death atlas of the postnatal mouse ventral forebrain and hypothalamus: effects of age and sex. J Comp Neurol 521:2551-69
Gilmore, Richard F; Varnum, Megan M; Forger, Nancy G (2012) Effects of blocking developmental cell death on sexually dimorphic calbindin cell groups in the preoptic area and bed nucleus of the stria terminalis. Biol Sex Differ 3:5
Holmes, Melissa M; Niel, Lee; Anyan, Jeff J et al. (2011) Effects of Bax gene deletion on social behaviors and neural response to olfactory cues in mice. Eur J Neurosci 34:1492-9
Murray, E K; Varnum, M M; Fernandez, J L et al. (2011) Effects of neonatal treatment with valproic acid on vasopressin immunoreactivity and olfactory behaviour in mice. J Neuroendocrinol 23:906-14
Semaan, Sheila J; Murray, Elaine K; Poling, Matthew C et al. (2010) BAX-dependent and BAX-independent regulation of Kiss1 neuron development in mice. Endocrinology 151:5807-17

Showing the most recent 10 out of 23 publications