Skip to main content Accessibility help
×
Hostname: page-component-8448b6f56d-cfpbc Total loading time: 0 Render date: 2024-04-16T15:43:08.875Z Has data issue: false hasContentIssue false

15 - Animal models of sex differences in non-reproductive brain functions

Published online by Cambridge University Press:  04 November 2009

George T. Taylor
Affiliation:
Behavioral Neuroscience Group University of Missouri – St. Louis 8001 Natural Bridge Rd. St. Louis, MO 63121 USA
Juergen Weiss
Affiliation:
Center for Biomedicine University of Heidelberg INF 347 D-69120 Heidelberg Germany
Frank Zimmermann
Affiliation:
Center for Biomedicine University of Heidelberg INF 347 D-69120 Heidelberg Germany
Turgut Tatlisumak
Affiliation:
Helsinki University Central Hospital
Marc Fisher
Affiliation:
University of Massachusetts Medical School
Get access

Summary

Overview

A neuroscientist embarking on a study of reproduction is eminently aware that an animal model chosen must consider gender differences and the endocrine factors determining them. Neuroscientists embarking on a study of function unrelated to reproduction may require the same consideration. It is now clear that gonadal hormones are important contributors to sex differences in a wide array of non-reproductive brain activities.

This conclusion has emerged from studies with animal models, sometimes unexpectedly when the experimental paradigm has included both males and females as subjects. Other findings of sex differences were observed first in human populations that suggested further study with animal models.

This review is a sampling of the remarkable variety of sex differences uncovered in adult brain function of which a neuroscientist may be unaware. A recent personal experience serves as an example. A respected colleague mentioned a new project to be conducted in her laboratory using an animal model of neuropathic pain. The interest was on a hypothesized acute attenuation of pain from treatments with a glutamate antagonist. Because the animal colony had an abundance of female rats, the subjects were to be groups of females. The assumption was that the topic had little relevance to reproduction and reproductive hormones, and any findings could be generalized equally to both sexes. Yet, there are reasons to question both assumptions. Both pain and glutamatergic pathways are influenced by sex hormones, and males and females may respond quite differently.

Type
Chapter
Information
Handbook of Experimental Neurology
Methods and Techniques in Animal Research
, pp. 239 - 256
Publisher: Cambridge University Press
Print publication year: 2006

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Breedlove SM, Hampson E. Sexual differentiation of the brain and behavior. In Becker, JB, Breedlove, SM, Crews, D, McCarthy, MM (eds.) Behavioral Endocrinology, 2nd edn. Cambridge, MA: MIT Press, 2002, pp. 75–116.Google Scholar
Payne AP. Gonadal hormones and the sexual differentiation of the nervous system: mechanisms and interactions. In Stone, TW (ed.) Neurotransmitters and Neuromodulators: Neuroactive Steroids. Boca Raton, FL: CRC Press, 1996, pp. 153–175.Google Scholar
Balthazart, J, Tlemcani, O, Ball, GF. Do sex differences in the brain explain sex differences in the hormonal induction of reproductive behavior? What 25 years of research on the Japanese quail tells us. Hormones Behav. 1996, 30: 627–661.CrossRefGoogle ScholarPubMed
McEwen, BS, Alves, SE. Estrogen actions in the central nervous system. Endocrine Rev. 1999, 20: 279–307.Google ScholarPubMed
Darlington, C. The Female Brain. New York: Taylor & Francis, 2002.CrossRefGoogle Scholar
Cooke, B, Hegstrom, CD, Villeneuve, LS, Breedlove, SM. Sexual differentiation of the vertebrate brain: principles and mechanisms. Frontiers Neuroendocrinol. 1999, 20: 323–362.Google Scholar
Tabibnia, G, Cooke, BM, Breedlove, SM. Sex difference and laterality in the volume of mouse dentate gyrus granule cell layer. Brain Res. 1999, 827: 41–45.CrossRefGoogle ScholarPubMed
Woolley, CS, McEwen, BS, Beatty, WW. Roles of estradiol and progesterone in regulation of hippocampal dendritic spine density during the estrous cycle in the rat. J. Comp. Neurol. 1993, 336: 293–306.CrossRefGoogle ScholarPubMed
Geschwind, N, Galaburda, A. Cerebral lateralization: biological mechanisms, associations and pathology. Arch. Neurol. 1985, 42: 428–459.CrossRefGoogle ScholarPubMed
Don Carlos, LL, Garcia-Ovejero, D, Sarkey, S, Garcia-Segura, LM, Azcoitia, I. Androgen receptor immunoreactivity in forebrain axons and dendrites in the rat. Endocrinology 2003, 144: 3632–3638.CrossRefGoogle Scholar
Lu, SF, McKenna, SE, Cologer-Clifford, A, Nau, EA, Simon, NG. Androgen receptor in mouse brain: sex differences and similarities in autoregulation. Endocrinology 1998, 139: 1594–1601.CrossRefGoogle ScholarPubMed
Zhang, JQ, Cai, WQ, Zhou, S, Su, BY. Distribution and differences of estrogen receptor beta immunoreactivity in the brain of adult male and female rats. Brain Res. 2002, 935: 73–80.CrossRefGoogle ScholarPubMed
Michael, RP, Clancy, AN, Zumpe, D. Distribution of androgen receptor-like immunoreactivity in the brains of cynomolgus monkeys. J. Neuroendocrinol. 1995, 7: 713–719.CrossRefGoogle ScholarPubMed
Majewska MD. Sex differences in brain morphology and pharmacodynamics. In Jensvold, MF, Halbreich, U, Hamilton, J (eds.) Psychopharmacology and Women. Washington, DC: American Psychiatric Press, 1996, pp. 73–83.Google Scholar
Di Paolo, T. Modulation of brain dopamine transmission by sex steroids. Revi. Neurosci. 1994, 5: 27–41.Google ScholarPubMed
Maswood, S, Truitt, W, Hotema, M, Caldarola-Pastuszka, M, Uphouse, L. Estrous cycle modulation of extracellular serotonin in mediobasal hypothalamus: role of the serotonin transporter and terminal autoreceptors. Brain Res. 1999, 831: 146–154.CrossRefGoogle ScholarPubMed
Sundblad, C, Eriksson, E. Reduced extracellular levels of serotonin in the amygdala of androgenized female rats. Eur. Neuropsychopharmacol. 1997, 7: 253–259.CrossRefGoogle ScholarPubMed
Purdy RH, Paul SM. Potentiation of GABAergic neurotransmission by steroids. In Baulieu, EE, Robel, P, Schumacher, M (eds.) Neurosteroids: A New Regulatory Function in the Nervous System. Totowa, NJ: Humana Press, 1999, pp. 143–153.CrossRefGoogle Scholar
Cyr, M, Calon, F, Morissette, M, Di Paolo, T. Estrogenic modulation of brain activity: implications for schizophrenia and Parkinson's disease. J. Psychiat. Neurosci. 2002, 27: 12–27.Google ScholarPubMed
Church, M, Subramanian, M. Cocaine's lethality increases during late gestation in the rat: a study of “critical periods” of exposure. Am. J. Obstet. Gynecol. 1997, 176: 901–906.CrossRefGoogle Scholar
Pesce, ME, Acevedo, X, Pinardi, G, Miranda, HF. Gender differences in diazepam withdrawal syndrome in mice. Pharmacol. Toxicol. 1994, 75: 353–355.CrossRefGoogle ScholarPubMed
Becker JB. Hormonal influences on sensorimotor function. In Becker, JB, Breedlove, SM, Crews, D, McCarthy, M (eds.) Behavioral Endocrinology, 2nd edn. Cambridge, MA: MIT Press, 2002, pp. 497–526.Google Scholar
Yilmaz, O, Kanit, L, Okur, BE, Pogun, S. Effects of nicotine on active avoidance learning in rats: sex differences. Behav. Pharmacol. 1997, 8: 253–260.Google ScholarPubMed
Perrotti, LI, Beck, KD, Luine, VN, Quinones, V. Progesterone and cocaine administration affect serotonin in the medial prefrontal cortex of ovariectomized rats. Neurosci. Lett. 2000, 291: 155–158.CrossRefGoogle ScholarPubMed
Suzuki, T, Koike, Y, Yanaura, S, George, FR, Meisch, RA. Sex differences in physical dependence on pentobarbital in four inbred strains of rats. Gen. Pharmacol. 1992, 23: 487–492.CrossRefGoogle ScholarPubMed
Hu, M, Becker, JB. Effects of sex and estrogen on behavioral sensitization to cocaine in rats. J. Neurosci. 2003, 23: 693–699.CrossRefGoogle ScholarPubMed
Nabeshima, T, Yamaguchi, K, Furukawa, H, Kameyama, T. Role of sex hormones in sex-dependent differences in phencyclidine induced stereotyped behaviors in rats. Eur. J. Pharmacol. 1984, 105: 197–206.CrossRefGoogle ScholarPubMed
Taylor, GT, Farr, S, Klinga, K, Weiss, J. Chronic fluoxetine suppresses circulating estrogen and the enhanced spatial learning of estrogen-restored ovariectomized female rats. Psychoneuroendocrinology 2004, 100: 1–10.Google Scholar
Webb, B, Burnett, PW, Walker, DW. Sex differences in ethanol-induced hypnosis and hypothermia in young Long-Evans rats. Alcoholism: Clin. Exp. Res. 2002, 26: 695–704.CrossRefGoogle ScholarPubMed
Albert, D, Jonik, R, Gorzalka, B, et al. Serum estradiol concentration required to maintain body weight, attractivity, proceptivity, and receptivity in the ovariectomized female rat. Physiol. Behav. 1991, 49: 225–231.CrossRefGoogle ScholarPubMed
Haider, S, Haleem, DJ. Decreases of brain serotonin following a food restriction schedule of 4 weeks in male and female rats. Med. Sci. Monit. 2000, 6: 1061–1067.Google ScholarPubMed
Leshner, AI. An Introduction to Behavioral Endocrinology. New York: Oxford University Press, 1978.Google Scholar
Perrot-Sinal, T, Ossenkopp, KP, Kavaliers, M. Influence of a natural stressor (predator odor) on locomotor activity in the meadow vole (Microtus pennsylvanicus): modulation by sex, reproductive condition and gonadal hormones. Psychoneuroendocrinology 2000, 25: 259–276.CrossRefGoogle ScholarPubMed
Gorman MR, Lee TM. Hormones and biological rhythms. In Becker, J B, Breedlove, S M, Crews, D, McCarthy, M M (eds.) Behavioral Endocrinology, 2nd edn. Cambridge, MA: MIT Press, 2002, pp. 451–494.Google Scholar
Mitsushima, D, Win-Shwe, TT, Kimura, F. Sexual dimorphism in the GABAergic control of gonadotropin release in intact rats. Neurosci. Res. 2003, 46: 399–405.CrossRefGoogle ScholarPubMed
Robaire, B, Bayly, S. Testicular signaling: incoming and outgoing messages. Ann. NY Acad. Sci. 1988, 56: 251–260.Google Scholar
Barrett, AC, Smith, ES, Picker, MJ. Capsaicin-induced hyperalgesia and mu-opioid-induced antihyperalgesia in male and female Fischer 344 rats. J. Pharmacol. Exp. Therap. 2003, 307: 237–245.CrossRefGoogle ScholarPubMed
Mogil, JS, Chesler, EJ, Wilson, SG, Juraska, JM, Sternberg, WF. Sex differences in thermal nociception and morphine antinociception in rodents depend on genotype. Neurosci. Biobehav. Rev. 2000, 24: 375–389.CrossRefGoogle ScholarPubMed
Whitacre, CC, Reingold, SC, O'Looney, PA. A gender gap in autoimmunity. Science 1999, 283: 1277–1278.CrossRefGoogle ScholarPubMed
Braude, S, Lacey, E, Tang-Martinez, Z, Taylor, GT. Stress, testosterone, and the immunoredistribution hypothesis. Behav. Ecol. 1999, 100: 201–210.Google Scholar
Williams CL. Hormones and cognition in nonhuman animals. In Becker, JB, Breedlove, SM, Crews, D, McCarthy, MM (eds.) Behavioral Endocrinology, 2nd edn. Cambridge, MA: MIT Press, 2002, pp. 527–578.Google Scholar
Galea, , Kavalis, M, Ossenkopp, K, Hampson, D. Gonadal hormone levels and spatial learning performance in the Morris water maze in male and female meadow voles: Microtus pennsylvanicus. Hormones Behav. 1995, 29: 106–125.CrossRefGoogle ScholarPubMed
Warren, SG, Juraska, JM. Spatial and nonspatial learning across the rat estrous cycle. Behav. Neurosci. 1997, 111: 259–266.CrossRefGoogle ScholarPubMed
Behl, C. Oestrogen as a neuroprotective hormone. Nature Rev. Neurosci. 2002, 3: 433–442.CrossRefGoogle ScholarPubMed
Hest, A, Haaren, F, Poll, N. Perseverative responding in male and female Wistar rat: effects of gonadal hormones. Hormones Behav. 1989, 23: 57–67.CrossRefGoogle ScholarPubMed
Lacreuse, A, Herndon, JG, Killiany, RJ, Rosene, DL, Moss, MB. Spatial cognition in rhesus monkeys: male superiority declines with age. Hormones Behav. 1999, 36: 70–76.CrossRefGoogle ScholarPubMed
Brinton, RD. Cellular and molecular mechanisms of estrogen regulation of memory function and neuroprotection against Alzheimer's disease: recent insights and remaining challenges. Learning Memory 2001, 8: 121–133.CrossRefGoogle ScholarPubMed
Webber, KM, Bowen, R, Casadesus, G, et al. Gonadotropins and Alzheimer's disease: the link between estrogen replacement therapy and neuroprotection. Acta Neurobiol. Exp. 2004, 64: 113–118.Google ScholarPubMed
Dribben, WH, Nemmers, BM, Nardi, AR, et al. Chronic but not acute estradiol treatment protects against the neurodegenerative effects of NMDA receptor antagonists. Endocrine 2003, 21: 53–58.CrossRefGoogle ScholarPubMed
Stevens, JR. Schizophrenia: reproductive hormones and the brain. Am. J. Psychiat. 2002, 159: 713–719.CrossRefGoogle Scholar
Weiss JM, Kilts CD. Animal models of depression and schizophrenia. In Schatzberg, A, Nemeroff, C (eds.) Textbook of Psychopharmacology, 2nd edn. Washington, DC: American Psychiatric Press, 1998, pp. 89–131.Google Scholar
Taylor, GT, Bardgett, M, Csernansky, J, et al. Neuroleptic influences on a lateralized behavioral bias in unoperated rats. Psychopharmacology 1999, 144: 30–37.CrossRefGoogle ScholarPubMed
Hafner, H, Behrens, S, Vry, J, Gattaz, W. An animal model for the effects of estradiol on dopamine-mediated behavior: implications for sex differences in schizophrenia. Psychiat. Res. 1991, 38: 125–134.CrossRefGoogle Scholar
Estrada-Camarena, E, Fernandez-Guasti, A, Lopez-Rubalcava, C. Antidepressant-like effect of different estrogenic compounds in the forced swimming test. Neuropsychopharmacology 2003, 28: 830–838.CrossRefGoogle ScholarPubMed
Handa, RJ, Burgess, LH, Kerr, JE, O'Keefe, J. Gonadal steroid hormone receptors and sex differences in the hypothalamo-pituitary-adrenal axis. Hormones Behav. 1994, 28: 464–476.CrossRefGoogle ScholarPubMed
Karandrea, D, Kittas, CK. Contribution of sex and cellular context in the regulation of brain corticosteroid receptors following restraint stress. Neuroendocrinology 2000, 71: 343–353.CrossRefGoogle ScholarPubMed
Alkayed, NJ, Harukuni, I, Kimes, AS, et al. Gender-linked brain injury in experimental stroke. Stroke 1998, 29: 159–165.CrossRefGoogle ScholarPubMed
Rusa, R, Alkayed, NJ, Crain, BJ, et al. 17beta-estradiol reduces stroke injury in estrogen-deficient female animals. Stroke 1999, 30: 1665–1670.CrossRefGoogle ScholarPubMed
Yang, SH, Perez, E, Cutright, J, et al. Testosterone increases neurotoxicity of glutamate in vitro and ischemia-reperfusion injury in an animal model. J. Appl. Physiol. 2002, 92: 195–201.CrossRefGoogle Scholar

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

Available formats
×