Elsevier

Brain Research

Volume 1366, 17 December 2010, Pages 233-245
Brain Research

Research Report
Sex differences in β-amyloid accumulation in 3xTg-AD mice: Role of neonatal sex steroid hormone exposure

https://doi.org/10.1016/j.brainres.2010.10.009Get rights and content

Abstract

The risk of Alzheimer's disease (AD) is higher in women than in men, a sex difference that likely results from the effects of sex steroid hormones. To investigate this relationship, we first compared progression of β-amyloid (Aβ) pathology in male and female triple transgenic (3xTg-AD) mice. We found that female 3xTg-AD mice exhibit significantly greater Aβ burden and larger behavioral deficits than age-matched males. Next, we evaluated how the organizational effects of sex steroid hormones during postnatal development may affect adult vulnerability to Aβ pathology. We observed that male 3xTg-AD mice demasculinized during early development exhibit significantly increased Aβ accumulation in adulthood. In contrast, female mice defeminized during early development exhibit a more male-like pattern of Aβ pathology in adulthood. Taken together, these results demonstrate significant sex differences in pathology in 3xTg-AD mice and suggest that these differences may be mediated by organizational actions of sex steroid hormones during development.

Research Highlights

►Female 3xTg-AD mice show accelerated Alzheimer-related pathology in comparison to male 3xTg-AD mice. ►Neonatally defeminized females exhibit a more male-like pattern of pathology. ►Neonatally demasculinized males exhibit a more female-like pattern of pathology. ►Sex differences in Alzheimer pathology involve organizational effects of sex steroid hormones.

Introduction

Women have a higher risk for the development of Alzheimer's disease (AD) than men. Epidemiological and observational studies have demonstrated a higher prevalence (Bachman et al., 1992, Jorm et al., 1987, Rocca et al., 1986) and incidence (Andersen et al., 1999, Fratiglioni et al., 2001, Jorm and Jolley, 1998, Ruitenberg et al., 2001) of AD in women. Further, AD appears to affect men and women differently, with women showing greater vulnerability to the disease. For example, at early stages of neurofibrillary tangle development, women exhibit greater senile plaque deposition than men (Corder et al., 2004). In addition, AD pathology is more strongly associated with clinical dementia in women than in men (Barnes et al., 2005).

Sex differences in AD may reflect differences between men and women in either of the two classic actions of sex steroid hormones: organizational effects during critical periods of development that induce permanent brain dimorphisms, or activational effects during adulthood that regulate adult brain function. Such activational effects of estrogens and testosterone are differentially diminished during aging in men and women. It is well established that estrogens have many beneficial effects in the brain, reviewed in (Wise, 2002) including numerous protective actions relevant to the prevention of AD, such as promotion of neuron viability and reduction of β-amyloid (Aβ) accumulation, reviewed in (Pike et al., 2009). The loss of neuroprotective estrogen actions as a consequence of estrogen depletion at menopause may increase the vulnerability of the female brain to AD and other disorders. Consistent with this theory, estrogen-based hormone therapy in postmenopausal women is associated with reduced risk of AD in some (Henderson et al., 1994, Kawas et al., 1997, Paganini-Hill and Henderson, 1996, Tang et al., 1996, Zandi et al., 2002) but not all studies (Espeland et al., 2004, Rapp et al., 2003). Similarly, ovariectomy-induced hormone depletion in wild-type rodents (Petanceska et al., 2000) and some transgenic mouse models of AD (Carroll et al., 2007, Levin-Allerhand et al., 2002, Zheng et al., 2002) can increase levels of the AD-related protein β-amyloid (Aβ), an effect attenuated by estradiol treatment. In men, age-related testosterone loss is linked with elevated levels of Aβ (Gillett et al., 2003, Rosario et al., 2009) and increased AD risk (Hogervorst et al., 2001, Moffat et al., 2004, Rosario et al., 2004, Rosario et al., 2009). In transgenic mouse models of AD, Aβ accumulation is accelerated under low testosterone conditions and reduced in the presence of high testosterone or dihydrotestosterone (McAllister et al., 2010, Rosario et al., 2006). Taken together, these studies suggest that adult exposure to estrogens and androgens may regulate the development of AD in women and men, respectively.

Another factor that may contribute to sex differences in AD risk is the organizational effects of sex steroid hormones during early development. Developmental patterns of estrogen and testosterone exposure induce numerous structural and functional differences between male and female brains (Cosgrove et al., 2007). Importantly, men and women also exhibit different vulnerabilities to several neurological disorders that occur prior to age-related hormone depletion, including post-traumatic stress disorder, schizophrenia, multiple sclerosis, autism, attention deficit disorder, and Tourette's syndrome (Cahill, 2006, Vagnerova et al., 2008). Thus, differences between male and female brains established during development may contribute to sex differences in vulnerability to disease, perhaps including AD.

The higher risk of AD in women may involve sex differences in the activational effects and/or organizational effects of estrogens and testosterone. To investigate these issues, we first compared the development of Aβ pathology in adult male and female triple transgenic (3xTg-AD) mice. Next, we assessed how the adult pattern of Aβ pathology is affected by disruption of the normal organizational effects of sex steroid hormones during critical developmental periods. Specifically, we defeminized neonatal female 3xTg-AD mice using transient testosterone treatment (Meek et al., 2006, Isgor and Sengelaub, 2003, Akhmadeev and Kalimullina, 2005) and demasculinized neonatal male 3xTg-AD mice by temporarily blocking testosterone action using the androgen receptor antagonist flutamide (Miyata et al., 2003, Houtsmuller et al., 1994, Meek et al., 2006, Kudwa et al., 2005). Alterations in adult pathology following these neonatal manipulations provide novel insight into the organizational role of sex steroid hormones on AD-related pathology.

Section snippets

Experiment 1: comparison of pathology development in male and female 3xTg-AD mice

To assess potential sex differences in AD-related pathology, Aβ accumulation and behavioral performance were compared in male and female 3xTg-AD mice across three age groups: 2–4 months, 6–8 months, and 12–14 months. The average age of males and females at each time point were not statistically different within age groups (average age in months ± standard error): female 2–4 months = 3.1 ± 0.0, male 2–4 months = 3.2 ± 0.1; (p = 0.78); female 6–8 months = 6.7 ± 0.3, male 6–8 months = 7.0 ± 0.4 (p = 0.37); female 12–14 months = 

Discussion

In this study, we investigated sex differences in the progression of Aβ pathology in male and female 3xTg-AD mice and how this relationship is affected by alterations in sex steroid hormones during a critical neonatal period of neural development and sexual differentiation. Consistent with our previous findings (Carroll et al., 2007, Rosario et al., 2006), we observed that Aβ accumulation and deficits in hippocampal-dependent behavior increase with age in adult male and female 3xTg-AD mice.

Experimental design

Male and female 3xTg-AD mice harboring three human transgenes, APP(Swe), PS1(M146V) and tau(P301L) (Oddo et al., 2003), were maintained at the USC Gerontology vivarium on 12 h light on/off cycle and given ad libitum access to food and water. Mice were handled in accordance with the NIH Health and Wellness of Animal Subjects procedures and an institutionally approved IACUC protocol.

Acknowledgments

This research was funded by NIH grants AG23739 (CJP) and AG026572 (RD Brinton/CJP). JCC was supported by NIH grant F31NS059174. ERR was supported by NIH grant NS52143. The authors thank Dr. Elizabeth Adkins-Regan and Dr. Ruth Wood for their insightful comments.

References (111)

  • S.J. Duff et al.

    A sex difference on a novel spatial working memory task in humans

    Brain Cogn.

    (2001)
  • A.C. Gore

    Developmental programming and endocrine disruptor effects on reproductive neuroendocrine systems

    Front. Neuroendocrinol.

    (2008)
  • C. Hirata-Fukae et al.

    Females exhibit more extensive amyloid, but not tau, pathology in an Alzheimer transgenic model

    Brain Res.

    (2008)
  • E.J. Houtsmuller et al.

    SDN-POA volume, sexual behavior, and partner preference of male rats affected by perinatal treatment with ATD

    Physiol. Behav.

    (1994)
  • C. Isgor et al.

    Prenatal gonadal steroids affect adult spatial behavior, CA1 and CA3 pyramidal cell morphology in rats

    Horm. Behav.

    (1998)
  • D.L. King et al.

    Behavioral characterization of the Tg2576 transgenic model of Alzheimer's disease through 19 months

    Physiol. Behav.

    (2002)
  • D.L. King et al.

    Progressive and gender-dependent cognitive impairment in the APP(SW) transgenic mouse model for Alzheimer's disease

    Behav. Brain Res.

    (1999)
  • L. Li et al.

    Augmented axonal defects and synaptic degenerative changes in female GRK5 deficient mice

    Brain Res. Bull.

    (2009)
  • S. Lutchmaya et al.

    2nd to 4th digit ratios, fetal testosterone and estradiol

    Early Hum. Dev.

    (2004)
  • M.D. Madeira et al.

    Sexual dimorphism in the mammalian limbic system

    Prog. Neurobiol.

    (1995)
  • C.M. McCormick et al.

    Persistent effects of prenatal, neonatal, or adult treatment with flutamide on the hypothalamic-pituitary-adrenal stress response of adult male rats

    Horm. Behav.

    (1999)
  • C.M. McCormick et al.

    Neonatal sex hormones have ‘organizational’ effects on the hypothalamic-pituitary-adrenal axis of male rats

    Brain Res. Dev. Brain Res.

    (1998)
  • L.R. Meek et al.

    Effects of prenatal stress on sexual partner preference in mice

    Physiol. Behav.

    (2006)
  • A. Mendrek

    Reversal of normal cerebral sexual dimorphism in schizophrenia: evidence and speculations

    Med. Hypotheses

    (2007)
  • P. Negri-Cesi et al.

    Metabolism of steroids in the brain: a new insight into the role of 5alpha-reductase and aromatase in brain differentiation and functions

    J. Steroid Biochem. Mol. Biol.

    (1996)
  • V.L. Nordell et al.

    Differential effects of estrogen in the injured forebrain of young adult and reproductive senescent animals

    Neurobiol. Aging

    (2003)
  • S. Oddo et al.

    Triple-transgenic model of Alzheimer's disease with plaques and tangles: intracellular Abeta and synaptic dysfunction

    Neuron

    (2003)
  • S.S. Petanceska et al.

    Ovariectomy and 17beta-estradiol modulate the levels of Alzheimer's amyloid beta peptides in brain

    Exp. Gerontol.

    (2000)
  • C.J. Pike et al.

    Protective actions of sex steroid hormones in Alzheimer's disease

    Front. Neuroendocrinol.

    (2009)
  • P.J. Pistell et al.

    Acquisition of conditioned taste aversion is impaired in the amyloid precursor protein/presenilin 1 mouse model of Alzheimer's disease

    Neuroscience

    (2008)
  • A. Ruitenberg et al.

    Incidence of dementia: does gender make a difference?

    Neurobiol. Aging

    (2001)
  • D.F. Swaab et al.

    Structural and functional sex differences in the human hypothalamus

    Horm. Behav.

    (2001)
  • M.X. Tang et al.

    Effect of oestrogen during menopause on risk and age at onset of Alzheimer's disease

    Lancet

    (1996)
  • A.V. Akhmadeev et al.

    Neonatal androgenization of female rats during sex differentiation of the brain modifies neuronal organization of the amygdala complex

    Ontogenez

    (2005)
  • N.J. Alkayed et al.

    Neuroprotective effects of female gonadal steroids in reproductively senescent female rats

    Stroke

    (2000)
  • K. Andersen et al.

    Gender differences in the incidence of AD and vascular dementia: the EURODEM studies

    Neurology

    (1999)
  • D.L. Bachman et al.

    Prevalence of dementia and probable senile dementia of the Alzheimer type in the Framingham study

    Neurology

    (1992)
  • L.L. Barnes et al.

    Sex differences in the clinical manifestations of Alzheimer disease pathology

    Arch. Gen. Psychiatry

    (2005)
  • A.M. Barron et al.

    The role of gonadotropins in Alzheimer's disease: potential neurodegenerative mechanisms

    Endocr.

    (2006)
  • D. Becu-Villalobos et al.

    Brain sexual differentiation and gonadotropins secretion in the rat

    Cell. Mol. Neurobiol.

    (1997)
  • S.M. Breedlove

    Neonatal androgen and estrogen treatments masculinize the size of motoneurons in the rat spinal nucleus of the bulbocavernosus

    Cell. Mol. Neurobiol.

    (1997)
  • L. Cahill

    Why sex matters for neuroscience

    Nat. Rev. Neurosci.

    (2006)
  • T. Canli et al.

    Sex differences in the neural basis of emotional memories

    Proc. Natl Acad. Sci. USA

    (2002)
  • J.C. Carroll et al.

    Selective estrogen receptor modulators differentially regulate Alzheimer-like changes in female 3xTg-AD mice

    Endocrinology

    (2008)
  • J.C. Carroll et al.

    Progesterone and estrogen regulate Alzheimer-like neuropathology in female 3xTg-AD mice

    J. Neurosci.

    (2007)
  • J.C. Carroll et al.

    Continuous and cyclic progesterone differentially interact with estradiol in the regulation of Alzheimer-like pathology in female 3xTransgenic-Alzheimer's disease mice

    Endocrinology

    (2010)
  • G. Casadesus et al.

    Evidence for the role of gonadotropin hormones in the development of Alzheimer disease

    Cell. Mol. Life Sci.

    (2005)
  • C.D. Conrad et al.

    Chronic stress impairs rat spatial memory on the Y maze, and this effect is blocked by tianeptine pretreatment

    Behav. Neurosci.

    (1996)
  • B.M. Cooke et al.

    Sexually dimorphic synaptic organization of the medial amygdala

    J. Neurosci.

    (2005)
  • E.H. Corder et al.

    The biphasic relationship between regional brain senile plaque and neurofibrillary tangle distributions: modification by age, sex, and APOE polymorphism

    Ann. NY Acad. Sci.

    (2004)
  • Cited by (195)

    View all citing articles on Scopus
    View full text