Article in press - uncorrected proof Horm Mol Biol Clin Invest 2011;7(3):385–391  2011 by Walter de Gruyter • Berlin • Boston. DOI 10.1515/HMBCI.2011.112

Sex differences in the injured brain

Roberto Cosimo Melcangi1,* and Luis M. GarciaSegura2 1

Department of Endocrinology, Pathophysiology and Applied Biology – Center of Excellence on Neurodegenerative Diseases, Universita` degli Studi di Milano, Milano, Italy 2 Instituto Cajal, C.S.I.C., Avenida Doctor Arce 37, Madrid, Spain

Abstract Observations obtained in human and in experimental models clearly demonstrate sex differences in degenerative events occurring in the central nervous system. The present review focuses on potential factors that may contribute to these sex-dimorphic features; in particular, morphological organization of the central nervous system and functional influence by neuroactive steroids, genes, and immune system are considered.

expression of specific genes in the adult central nervous system. Sex differences in the levels of steroid hormones, neurosteroids (i.e., steroids synthesized in the nervous system), and neuroactive steroids (i.e., a definition including both steroid hormones and neurosteroids) in adulthood is the factor that has been explored in more detail, since there is solid evidence indicating that these hormones may affect the outcome of insults to the brain and the spinal cord. Finally, sex differences in the immune system have been reported. This is a critical factor for the manifestation of sex differences in autoimmune diseases and may also be relevant for other inflammatory conditions. We will briefly review here some representative examples of sex differences in brain pathology, with information obtained from clinical and animal studies. Then we will analyze the potential influence of the factors mentioned above in the generation of these differences.

Sex differences in brain pathology Keywords: gender; inflammation; neuroactive steroids; neurodegenerative diseases.

Introduction Clinical evidence and studies in animal models of central nervous system injury and neurodegeneration have revealed the existence of sex differences in the pathology of the brain and the spinal cord. These differences may be manifested in the age of onset, incidence, prevalence, or prognosis of the disease, in the tissue damage associated with the pathological condition, and/or in the response to rehabilitation or neuroprotective therapies. There are several potential factors that may contribute to generate sex differences in brain pathology. One of these factors is the existence of sex differences in the functional and morphological organization of the central nervous system. These differences are generated during the developmental period and may be the direct consequence of the expression by neural cells of specific sex differentiation genes. In addition, steroid milieu regulates the process of sex differentiation of the brain and the spinal cord. Another potential source for sex differences in brain pathology, although less explored, is the sexually dimorphic *Corresponding author: Roberto Cosimo Melcangi, Department of Endocrinology, Pathophysiology and Applied Biology – Center of Excellence on Neurodegenerative Diseases, Universita` degli Studi di Milano, Via Balzaretti 9, 20133 Milano, Italy Phone: q39-02-50318238, Fax: q39-02-50318204, E-mail: [email protected] Received May 28, 2011; accepted July 6, 2011; previously published online August 15, 2011

Sex differences in the incidence and manifestation of neurodegenerative diseases

Studies in different animal models of neurodegenerative diseases have revealed the existence of sex differences in brain damage w1x. In some cases females are more affected by the pathology than males. For instance, female mice show higher seizure activity and more hippocampal neurodegeneration than male mice after the administration of kainic acid, a model of epilepsy and of excitotoxic neurodegeneration w2x. In addition, females show higher plaque load than males in different amyloid precursor protein (APP) transgenic animal models of Alzheimer’s disease w3, 4x. In transgenic mice expressing mutant APP, preselinin-1, and tau, female animals show an increased cognitive impairment w5x and an increased production and decreased degradation of b-amyloid compared to males w6x. In contrast, in a model of Parkinson’s disease in mice, caused by the intoxication with 1-methyl4-phenyl-1,2,3,6-tetrahydropyridine, male animals show a greater depletion of dopamine than female animals w7x. Also, in transgenic animal models of Huntington’s disease, males show greater motor deficits and increased loss and atrophy of dopaminergic neurons than females w8, 9x. Sex differences in the incidence and manifestation of neurodegenerative diseases have been observed in humans w1x. The incidence of Parkinson’s disease is greater in men than in women w10–13x, and women present higher age at onset and milder motor deterioration than men w14x. Age of onset of Huntington’s disease is also higher and the course of the illness more moderate in women than in men w15–17x. In contrast, epidemiological studies support a higher prevalence

2011/0112

Article in press - uncorrected proof 386 Melcangi and Garcia-Segura: Sex dimorphic features in the brain

and incidence of Alzheimer’s disease in women w18–21x. Sex differences in Alzheimer’s disease patients are also evidenced in terms of plaque load, with a significantly different distribution of them in cortical areas of female patients w22x. Women also show more neurofibrillary tangles w23x. Analysis of visuospatial episodic memory also shows sex differences, with a better performance in male than in female Alzheimer’s disease patients w24x. Clinical manifestation of dementia among Alzheimer’s disease patients is more common in women than in men w23x. As it has been reported for other autoimmune diseases w25x, the incidence of multiple sclerosis is higher in women than in men w26–28x. However, men have a worse prognosis than women w29x. Indeed, men are affected in older age and develop a more severe pathology, defined as a shorter time to reach severe disability w30x. Cognitive decline is also more predominant in men than in women w31, 32x. Sex differences in the incidence and outcome of stroke

Stroke is another pathological condition for which sex differences in the underlying etiology, presentation, and outcome have been reported w1, 33, 34x. In experimental stroke, young female animals show less damage than young males w35–39x. However, this sex difference is not observed in older individuals w40x. Human studies indicate that the incidence of stroke is higher in men than in women. However, after 85 years of age, there is a higher incidence of stroke in women w41x, which may be in part due to the fact that women have strokes at older ages than men. In addition, women have a poorer recovery from stroke than men w33, 34, 42–45x. However, while some studies suggest increased long-term mortality in women vs. men after stroke, other studies indicate the opposite w33, 34, 41, 46–48x. Sex differences in the outcome of traumatic brain injury

Some observations indicate that mortality and poor outcomes (i.e., severe disability or persistent vegetative state) after traumatic brain injury were significantly higher in females than in males with the same extent of injury w49–51x; however, others seem to suggest the opposite. Thus, females have better overall responses to rehabilitation therapy w52x. Moreover, as indicated by the Glasgow Coma Scale scores and length of post-traumatic amnesia, men have greater level of injury w53x. Similar results were obtained using the Wisconsin card sort test as a measure of executive function w54x. Furthermore, 1 year from injury women show better performance on cognitive outcome measures w55x. Finally, further observations indicate that, after moderate-to-severe traumatic brain injury, outcomes are significantly worse in age-matched males than in post-menopausal females. This difference is not observed when males and pre-menopausal females are compared w56x. Interesting observations have also been obtained in cerebral spinal fluid of patients. These suggest that females have smaller oxidative damage loads than males w57x, and a significant sex difference also occurs

in glutamate and lactate/pyruvate production w58x and in lipid peroxidation w59x.

Possible causes of sex differences in brain pathology Sexual differentiation of the nervous system

Sex differences in brain pathology may be the consequence of the morphological and functional differences in neural structure between the sexes w60x. Both genetic sex and developmental actions of sex hormones contribute to the generation of these sex differences w61–63x and both factors may therefore influence the risk and outcome of neurodegenerative and neurological diseases w1, 64, 65x. The importance of genetically-determined or hormonally-generated sex differences in neural cells in the sexually dimorphic response of the nervous system to injury is suggested by the existence of sex differences in the in vitro response of neurons, glial cells, and brain slices to damaging agents w66–69x. In addition, stress hormones during the developmental period may affect brain differentiation and may have permanent effects on brain function. Thus, pre-natal stress and maternal separation have been reported to affect the structure and function of different brain regions w70–72x. The alterations caused by pre-natal and early post-natal stress may contribute to facilitate brain deterioration with aging w72x. Interestingly, pre-natal and early post-natal stress have different consequences in males and females w70, 71, 73x, and may contribute to the generation of sex differences in brain pathology. Sex differences in gene expression in adulthood

In addition to the influence of sex determination genes in the differentiation of the brain, the expression of genes located in the Y chromosome during adulthood w74x is a potential source of sex differences in brain pathology. Furthermore, the partial inactivation of X-linked chromosome genes in females w75x may also generate differences in brain gene expression during adult life w74x that, in turn, may influence the response of neural tissue to injury and degeneration w76x. The influence of the steroid milieu in adulthood

Neuroactive steroids exert neuroprotective actions w1, 77–83x. Therefore, changes in the levels of sex steroid hormones during the ovarian cycle and aging may contribute to the generation of sex differences in the manifestation of brain pathology. Indeed, in female rodents it has been demonstrated that the fluctuation in hormonal levels during the estrous cycle affects the response of the brain to pathological insults. For instance, the neurotoxic effect of kainic acid on hippocampal neurons in intact female rats is different depending on the day of the estrous cycle on which the neurotoxin is injected. No significant neuronal loss is observed when a moderate dose of kainic acid is injected on the morning of estrus, 1 day after the peak of estrogen levels in plasma. In

Article in press - uncorrected proof Melcangi and Garcia-Segura: Sex dimorphic features in the brain

contrast, there is a significant loss of hilar neurons when the same dose of kainic acid is injected in the morning of proestrus, before the peak of estrogen levels in plasma, as well as when it is injected into ovariectomized rats w84x. Both circulating testosterone and estradiol as well as estradiol locally synthesized within the brain have been shown to exert neuroprotective effects in this experimental model of neurodegeneration w85x. Animals injected with kainic acid may develop seizures and, as we have mentioned previously, the administration of this neurotoxin to rodents is often used as a model of epilepsy. Interestingly, seizures involving the limbic system are also influenced by the menstrual cycle in epileptic women w65x; seizure activity incidence is low when progesterone levels in plasma are high compared to estradiol levels. In contrast, seizure activity is higher when both progesterone and estradiol are at low levels in plasma and during the follicular phase, when there is an abrupt increase in estradiol plasma levels w65, 86x. Sex steroid hormones may be involved in sex differences in the outcome of experimental stroke, because the damage in female animals is enhanced by ovariectomy w39x. However, the balance of androgens and estrogens may also determine sex differences in the outcome of neurodegeneration since androgens enhance brain damage induced by middle cerebral artery occlusion in male animals w87–89x. This may also be the case in other pathologies. For instance, decreased levels of estradiol correlate with an increased neurodegeneration in males in an experimental rat model of Huntington disease w8x. The balance of estrogens and androgens seems also to determine the sexual dimorphism on the neurodegenerative effect of the non-competitive NMDA receptorantagonist MK801 in the granular retrosplenial cortex of rats, which is more manifest in females w90x. In this case, however, androgens seem to inhibit and estradiol seems to increase neurodegeneration. In a model of Parkinson’s disease, caused by the lesion of the nigrostriatal dopaminergic pathway with 6-hydroxydopamine, estrogen is neuroprotective in females but not in males w91, 92x. However, local production of estradiol within the brain appears to protect against neurotoxin-induced striatal damage in both male and female rodents w93, 94x. Sex differences in the response of the striatal dopaminergic system to estradiol may be in part the consequence of sex differences in the structural and functional organization of the dopaminergic synaptic circuits w91x. However, the causes for the different outcome of central and peripheral estradiol in males remain to be elucidated. On the other hand, neuronal damage in female rodents in experimental models of Parkinson’s disease is influenced by the endogenous fluctuation of gonadal hormones during the estrous cycle w91, 95–97x. In addition, regulation of estrogen receptor alpha turnover by the Parkinson’s disease-related protein parkin may also be involved in the sex difference in this neurodegenerative disease w98x. Therefore, both sex differences in the organization of the nigrostriatal dopaminergic system, sex differences in circulating hormones, and sex differences in the sensitivity and response of neural tissue to these hormones may contribute to determine sex differences in the manifestation of the pathology.

387

It is also important to consider that brain injury affects the levels of neuroactive steroids in the nervous system and plasma w99, 100x. The regulation of the levels of neuroactive steroids after neural injury may present sexually dimorphic profiles and may be affected by the endocrine status of the animals w101–104x. Sex differences in brain steroid levels after injury may be the consequence of sex differences in the expression and/or activity of steroidogenic enzymes in the nervous system. For instance, sex differences in the expression and activity of the enzyme aromatase in astrocytes has been reported. This finding suggests that astrocytes from female brains produce more estradiol than astrocytes from males w69x. Because aromatase expression in astrocytes is induced after brain injury and is neuroprotective w80–82, 85x, the reported sex differences in the expression of aromatase by astrocytes may be relevant to explain sex differences in the manifestation of brain pathologies and to explain sex differences in the response of the injured male and female brain to estradiol therapy. In addition to sex differences in peripheral or central steroid levels, sex differences in the response of neurons and glial cells to hormonal steroids may also be involved in the manifestation of sex differences in brain pathology. Sex differences in the action of glucocorticoids in the brain may contribute to the generation of sex differences in brain damage. It has been proposed that the female brain has a different innate strategy to handle stress than the male brain w73, 105x. An enhanced corticosterone response in females may be the cause of sex differences in cognitive performance in an experimental model of Alzheimer’s disease w5x. Neurons and glial cells also show a sexually dimorphic response to gonadal hormones. For instance, repeated estradiol pulses modulate hippocampal neurogenesis and neuronal death in adult female rodents but not in adult male rodents, while testosterone and dihydrotestosterone upregulate hippocampal neurogenesis in adult male rodents w106x. In addition, gonadal hormones have different effects in the survival of oligodendrocytes in cultures from female mice compared to cultures from male mice w107x. Estradiol has more potent effects in decreasing proliferation and increasing cell death in primary cortical astrocytic cultures from females than in astrocytes from males w108x. These differences may be highly relevant for demyelinating diseases and other pathological conditions in which white matter is affected. The differential effects of glucocorticoids and sex steroid hormones in male and female neural cells may be a consequence of the existence of sex differences in the levels of steroid receptors or in the mechanisms of steroid signaling. For instance, progesterone, estradiol, and dihydrotestosterone have different effects in the activation of Akt in male and female oligodendrocytes w107x and estradiol has different effects on the activation of ERK1 and ERK2 in male and female astrocytes w108x. In addition, male and female brains may respond to injury with different changes in the expression of steroid receptors. Thus, sex differences in the damage produced in the cerebral cortex by middle cerebral artery occlusion may be associated with sex differences in the regulation of estrogen receptor alpha expression, which is

Article in press - uncorrected proof 388 Melcangi and Garcia-Segura: Sex dimorphic features in the brain

increased in the cerebral cortex of female rats, but not in males, after the brain injury w109x. 2.

The influence of the immune system

Sex differences in immune responses w110x, including sex differences in CNS immunity w111x, may contribute to different outcome of pathological insults to the brain and the spinal cord. Sex differences in pro-inflammatory responses, which have been described in multiple sclerosis patients w112, 113x and in experimental autoimmune encephalomyelitis w114–117x, may contribute to the sex differences in the manifestation of the pathology. Indeed, MRI analyses have shown that men develop less inflammatory, but more destructive, lesions than women w118x. Sex differences in the inflammatory response also contribute to the generation of sex differences in brain damage in experimental models of Alzheimer’s w119x, Parkinson’s w120x, and cerebrovascular disease w121x. Both glucocorticoids and sex steroid hormones are known to regulate the inflammatory responses of astrocytes, microglia, and peripheral immune cells w110, 122, 123x, and may therefore be involved in the generation of sex differences in CNS immunity.

3.

4.

5.

6.

7.

Concluding remarks 8.

We have here reviewed evidence indicating that the pathological manifestations of the central nervous system after injury and under neurodegenerative conditions present sexually dimorphic traits. We have also analyzed some potential causes for these differences, in particular the influence of genetic, endocrine, and immune factors. However, there are other variables that have not been reviewed here, such as sex differences in behavior, which are not always associated to reproduction. It is becoming evident that physical exercise and other factors associated to individual’s attitudes, including diet and drug consumption, may affect cognition and the outcome of brain pathology. In humans, cultural factors that impose different behavioral patterns in men and women may result in gender differences in life style, which in turn may generate gender differences in the risk of neurodegenerative diseases and brain injury. These and other factors should be further explored to understand the causes of sex differences in brain pathology, with the final aim of developing genderspecific strategies to prevent and counteract central nervous system damage.

9.

10.

11.

12.

13.

Acknowledgements The financial support of Fondazione San Paolo (Progetto Neuroscienze, PF-2009.1180), Fondazione Italiana Sclerosi Multipla (2010/ 23) and PUR from Universita degli Studi di Milano, Italy, to R.C. Melcangi is gratefully acknowledged.

References 1. Melcangi RC, Garcia-Segura LM. Sex-specific therapeutic strategies based on neuroactive steroids: in search for

14.

15.

16.

innovative tools for neuroprotection. Horm Behav 2010;57: 2–11. Zhang XM, Zhu SW, Duan RS, Mohammed AH, Winblad B, Zhu J. Gender differences in susceptibility to kainic acidinduced neurodegeneration in aged C57BL/6 mice. Neurotoxicology 2008;29:406–12. Callahan MJ, Lipinski WJ, Bian F, Durham RA, Pack A, Walker LC. Augmented senile plaque load in aged female betaamyloid precursor protein-transgenic mice. Am J Pathol 2001;158:1173–7. Lewis J, Dickson DW, Lin WL, Chisholm L, Corral A, Jones G, Yen SH, Sahara N, Skipper L, Yager D, Eckman C, Hardy J, Hutton M, McGowan E. Enhanced neurofibrillary degeneration in transgenic mice expressing mutant tau and APP. Science 2001;293:1487–91. Clinton LK, Billings LM, Green KN, Caccamo A, Ngo J, Oddo S, McGaugh JL, LaFerla FM. Age-dependent sexual dimorphism in cognition and stress response in the 3xTg-AD mice. Neurobiol Dis 2007;28:76–82. Hirata-Fukae C, Li HF, Hoe HS, Gray AJ, Minami SS, Hamada K, Niikura T, Hua F, Tsukagoshi-Nagai H, Horikoshi-Sakuraba Y, Mughal M, Rebeck GW, LaFerla FM, Mattson MP, Iwata N, Saido TC, Klein WL, Duff KE, Aisen PS, Matsuoka Y. Females exhibit more extensive amyloid, but not tau, pathology in an Alzheimer transgenic model. Brain Res 2008;1216:92–103. Czlonkowska A, Ciesielska A, Gromadzka G, KurkowskaJastrzebska I. Gender differences in neurological disease: role of estrogens and cytokines. Endocrine 2006;29:243–56. Bode FJ, Stephan M, Suhling H, Pabst R, Straub RH, Raber KA, Bonin M, Nguyen HP, Riess O, Bauer A, Sjoberg C, Petersen A, von Horsten S. Sex differences in a transgenic rat model of Huntington’s disease: decreased 17beta-estradiol levels correlate with reduced numbers of DARPP32q neurons in males. Hum Mol Genet 2008;17:2595–609. Orr AL, Huang S, Roberts MA, Reed JC, Li S, Li XJ. Sexdependent effect of BAG1 in ameliorating motor deficits of Huntington disease transgenic mice. J Biol Chem 2008;283: 16027–36. Benito-Leon J, Bermejo-Pareja F, Rodriguez J, Molina JA, Gabriel R, Morales JM. Prevalence of PD and other types of Parkinsonism in three elderly populations of central Spain. Mov Disord 2003;18:267–74. de Lau LM, Giesbergen PC, de Rijk MC, Hofman A, Koudstaal PJ, Breteler MM. Incidence of Parkinsonism and Parkinson disease in a general population: the Rotterdam study. Neurology 2004;63:1240–4. Van Den Eeden SK, Tanner CM, Bernstein AL, Fross RD, Leimpeter A, Bloch DA, Nelson LM. Incidence of Parkinson’s disease: variation by age, gender, and race/ethnicity. Am J Epidemiol 2003;157:1015–22. Wooten GF, Currie LJ, Bovbjerg VE, Lee JK, Patrie J. Are men at greater risk for Parkinson’s disease than women? J Neurol Neurosurg Psychiatry 2004;75:637–9. Haaxma CA, Bloem BR, Borm GF, Oyen WJ, Leenders KL, Eshuis S, Booij J, Dluzen DE, Horstink MW. Gender differences in Parkinson’s disease. J Neurol Neurosurg Psychiatry 2007;78:819–24. Foroud T, Gray J, Ivashina J, Conneally PM. Differences in duration of Huntington’s disease based on age at onset. J Neurol Neurosurg Psychiatry 1999;66:52–6. Pekmezovic T, Svetel M, Maric J, Dujmovic-Basuroski I, Dragasevic N, Keckarevic M, Romac S, Kostic VS. Survival of Huntington’s disease patients in Serbia: longer survival in female patients. Eur J Epidemiol 2007;22:523–6.

Article in press - uncorrected proof Melcangi and Garcia-Segura: Sex dimorphic features in the brain

17. Roos RA, Vegter-van der Vlis M, Hermans J, Elshove HM, Moll AC, van de Kamp JJ, Bruyn GW. Age at onset in Huntington’s disease: effect of line of inheritance and patient’s sex. J Med Genet 1991;28:515–9. 18. Andersen K, Launer LJ, Dewey ME, Letenneur L, Ott A, Copeland JR, Dartigues JF, Kragh-Sorensen P, Baldereschi M, Brayne C, Lobo A, Martinez-Lage JM, Stijnen T, Hofman A. Gender differences in the incidence of AD and vascular dementia: the EURODEM Studies. EURODEM Incidence Research Group Neurology 1999;53:1992–7. 19. Fratiglioni L, Viitanen M, von Strauss E, Tontodonati V, Herlitz A, Winblad B. Very old women at highest risk of dementia and Alzheimer’s disease: incidence data from the Kungsholmen Project, Stockholm. Neurology 1997;48:132–8. 20. Azad NA, Al Bugami M, Loy-English I. Gender differences in dementia risk factors. Gend Med 2007;4:120–9. 21. Vina J, Lloret A, Valles SL, Borras C, Badia MC, Pallardo FV, Sastre J, Alonso MD. Effect of gender on mitochondrial toxicity of Alzheimer’s Abeta peptide. Antioxid Redox Signal 2007;9:1677–90. 22. Kraszpulski M, Soininen H, Helisalmi S, Alafuzoff I. The load and distribution of beta-amyloid in brain tissue of patients with Alzheimer’s disease. Acta Neurol Scand 2001;103:88–92. 23. Barnes LL, Wilson RS, Bienias JL, Schneider JA, Evans DA, Bennett DA. Sex differences in the clinical manifestations of Alzheimer disease pathology. Arch Gen Psychiatry 2005;62: 685–91. 24. Beinhoff U, Tumani H, Brettschneider J, Bittner D, Riepe MW. Gender-specificities in Alzheimer’s disease and mild cognitive impairment. J Neurol 2008;255:117–22. 25. Whitacre CC. Sex differences in autoimmune disease. Nat Immunol 2001;2:777–80. 26. Noonan CW, Kathman SJ, White MC. Prevalence estimates for MS in the United States and evidence of an increasing trend for women. Neurology 2002;58:136–8. 27. Orton SM, Herrera BM, Yee IM, Valdar W, Ramagopalan SV, Sadovnick AD, Ebers GC. Sex ratio of multiple sclerosis in Canada: a longitudinal study. Lancet Neurol 2006;5:932–6. 28. Schwendimann RN, Alekseeva N. Gender issues in multiple sclerosis. Int Rev Neurobiol 2007;79:377–92. 29. Hawkins SA, McDonnell GV. Benign multiple sclerosis? Clinical course, long term follow up, and assessment of prognostic factors. J Neurol Neurosurg Psychiatry 1999;67:148–52. 30. Confavreux C, Vukusic S, Adeleine P. Early clinical predictors and progression of irreversible disability in multiple sclerosis: an amnesic process. Brain 2003;126:770–82. 31. Savettieri G, Messina D, Andreoli V, Bonavita S, Caltagirone C, Cittadella R, Farina D, Fazio MC, Girlanda P, Le Pira F, Liguori M, Lugaresi A, Nocentini U, Reggio A, Salemi G, Tedeschi G, Trojano M, Valentino P, Quattrone A. Gender-related effect of clinical and genetic variables on the cognitive impairment in multiple sclerosis. J Neurol 2004;251:1208–14. 32. Parmenter BA, Denney DR, Lynch SG, Middleton LS, Harlan LM. Cognitive impairment in patients with multiple sclerosis: association with the APOE gene and promoter polymorphisms. Mult Scler 2007;13:25–32. 33. Turtzo LC, McCullough LD. Sex differences in stroke. Cerebrovasc Dis 2008;26:462–74. 34. Reeves MJ, Bushnell CD, Howard G, Gargano JW, Duncan PW, Lynch G, Khatiwoda A, Lisabeth L. Sex differences in stroke: epidemiology, clinical presentation, medical care, and outcomes. Lancet Neurol 2008;7:915–26.

389

35. Zhang YQ, Shi J, Rajakumar G, Day AL, Simpkins JW. Effects of gender and estradiol treatment on focal brain ischemia. Brain Res 1998;784:321–4. 36. Hall ED, Pazara KE, Linseman KL. Sex differences in postischemic neuronal necrosis in gerbils. J Cereb Blood Flow Metab 1991;11:292–8. 37. Alkayed NJ, Harukuni I, Kimes AS, London ED, Traystman RJ, Hurn PD. Gender-linked brain injury in experimental stroke. Stroke 1998;29:159–65. 38. Bramlett HM. Sex differences and the effect of hormonal therapy on ischemic brain injury. Pathophysiology 2005;12:17–27. 39. McCullough LD, Blizzard K, Simpson ER, Oz OK, Hurn PD. Aromatase cytochrome P450 and extragonadal estrogen play a role in ischemic neuroprotection. J Neurosci 2003;23:8701–5. 40. Alkayed NJ, Murphy SJ, Traystman RJ, Hurn PD, Miller VM. Neuroprotective effects of female gonadal steroids in reproductively senescent female rats. Stroke 2000;31:161–8. 41. Rosamond W, Flegal K, Friday G, Furie K, Go A, Greenlund K, Haase N, Ho M, Howard V, Kissela B, Kittner S, LloydJones D, McDermott M, Meigs J, Moy C, Nichol G, O’Donnell CJ, Roger V, Rumsfeld J, Sorlie P, Steinberger J, Thom T, Wasserthiel-Smoller S, Hong Y. Heart disease and stroke statistics – 2007 update: a report from the American Heart Association Statistics Committee and Stroke Statistics Subcommittee. Circulation 2007;115:e69–171. 42. Roquer J, Campello AR, Gomis M. Sex differences in firstever acute stroke. Stroke 2003;34:1581–5. 43. Niewada M, Kobayashi A, Sandercock PA, Kaminski B, Czlonkowska A. Influence of gender on baseline features and clinical outcomes among 17,370 patients with confirmed ischaemic stroke in the International Stroke Trial. Neuroepidemiology 2005;24:123–8. 44. Kapral MK, Fang J, Hill MD, Silver F, Richards J, Jaigobin C, Cheung AM. Sex differences in stroke care and outcomes: results from the Registry of the Canadian Stroke Network. Stroke 2005;36:809–14. 45. Holroyd-Leduc JM, Kapral MK, Austin PC, Tu JV. Sex differences and similarities in the management and outcome of stroke patients. Stroke 2000;31:1833–7. 46. Kimura K, Minematsu K, Kazui S, Yamaguchi T. Mortality and cause of death after hospital discharge in 10,981 patients with ischemic stroke and transient ischemic attack. Cerebrovasc Dis 2005;19:171–8. 47. Eriksson M, Norrving B, Terent A, Stegmayr B. Functional outcome 3 months after stroke predicts long-term survival. Cerebrovasc Dis 2008;25:423–9. 48. Devroey D, Van Casteren V, Buntinx F. Registration of stroke through the Belgian sentinel network and factors influencing stroke mortality. Cerebrovasc Dis 2003;16:272–9. 49. Ng I, Lee KK, Lim JH, Wong HB, Yan XY. Investigating gender differences in outcome following severe traumatic brain injury in a predominantly Asian population. Br J Neurosurg 2006;20:73–8. 50. Kraus JF, Peek-Asa C, McArthur D. The independent effect of gender on outcomes following traumatic brain injury: a preliminary investigation. Neurosurg Focus 2000;8:e5. 51. Farace E, Alves WM. Do women fare worse: a metaanalysis of gender differences in traumatic brain injury outcome. J Neurosurg 2000;93:539–45. 52. Groswasser Z, Cohen M, Keren O. Female TBI patients recover better than males. Brain Inj 1998;12:805–8. 53. Slewa-Younan S, Green AM, Baguley IJ, Gurka JA, Marosszeky JE. Sex differences in injury severity and outcome meas-

Article in press - uncorrected proof 390 Melcangi and Garcia-Segura: Sex dimorphic features in the brain

54.

55.

56.

57.

58.

59.

60.

61.

62.

63. 64. 65. 66. 67.

68.

69.

70.

71.

ures after traumatic brain injury. Arch Phys Med Rehabil 2004;85:376–9. Niemeier JP, Marwitz JH, Lesher K, Walker WC, Bushnik T. Gender differences in executive functions following traumatic brain injury. Neuropsychol Rehabil 2007;17:293–313. Ratcliff JJ, Greenspan AI, Goldstein FC, Stringer AY, Bushnik T, Hammond FM, Novack TA, Whyte J, Wright DW. Gender and traumatic brain injury: do the sexes fare differently? Brain Inj 2007;21:1023–30. Davis DP, Douglas DJ, Smith W, Sise MJ, Vilke GM, Holbrook TL, Kennedy F, Eastman AB, Velky T, Hoyt DB. Traumatic brain injury outcomes in pre- and post-menopausal females versus age-matched males. J Neurotrauma 2006;23:140–8. Wagner AK, Bayir H, Ren D, Puccio A, Zafonte RD, Kochanek PM. Relationships between cerebrospinal fluid markers of excitotoxicity, ischemia, and oxidative damage after severe TBI: the impact of gender, age, and hypothermia. J Neurotrauma 2004; 21:125–36. Wagner AK, Fabio A, Puccio AM, Hirschberg R, Li W, Zafonte RD, Marion DW. Gender associations with cerebrospinal fluid glutamate and lactate/pyruvate levels after severe traumatic brain injury. Crit Care Med 2005;33:407–13. Bayir H, Marion DW, Puccio AM, Wisniewski SR, Janesko KL, Clark RS, Kochanek PM. Marked gender effect on lipid peroxidation after severe traumatic brain injury in adult patients. J Neurotrauma 2004;21:1–8. Stein DG. Sex differences in brain damage and recovery of function: experimental and clinical findings. Prog Brain Res 2007;161:339–51. Reisert I, Pilgrim C. Sexual differentiation of monoaminergic neurons – genetic or epigenetic? Trends Neurosci 1991;14: 468–73. Arnold AP, Xu J, Grisham W, Chen X, Kim YH, Itoh Y. Minireview: sex chromosomes and brain sexual differentiation. Endocrinology 2004;145:1057–62. Arnold AP, Burgoyne PS. Are XX and XY brain cells intrinsically different? Trends Endocrinol Metab 2004;15:6–11. Lang JT, McCullough LD. Pathways to ischemic neuronal cell death: are sex differences relevant? J Transl Med 2008;6:33. Vagnerova K, Koerner IP, Hurn PD. Gender and the injured brain. Anesth Analg 2008;107:201–14. Li H, Pin S, Zeng Z, Wang MM, Andreasson KA, McCullough LD. Sex differences in cell death. Ann Neurol 2005;58:317–21. Du L, Bayir H, Lai Y, Zhang X, Kochanek PM, Watkins SC, Graham SH, Clark RS. Innate gender-based proclivity in response to cytotoxicity and programmed cell death pathway. J Biol Chem 2004;279:38563–70. Heyer A, Hasselblatt M, von Ahsen N, Hafner H, Siren AL, Ehrenreich H. In vitro gender differences in neuronal survival on hypoxia and in 17beta-estradiol-mediated neuroprotection. J Cereb Blood Flow Metab 2005;25:427–30. Liu M, Hurn PD, Roselli CE, Alkayed NJ. Role of P450 aromatase in sex-specific astrocytic cell death. J Cereb Blood Flow Metab 2007;27:135–41. Viveros MP, Llorente R, Lopez-Gallardo M, Suarez J, Bermudez-Silva F, De la Fuente M, Rodriguez de Fonseca F, GarciaSegura LM. Sex-dependent alterations in response to maternal deprivation in rats. Psychoneuroendocrinology 2009;34:S217– 26. Llorente R, Gallardo ML, Berzal AL, Prada C, Garcia-Segura LM, Viveros MP. Early maternal deprivation in rats induces gender-dependent effects on developing hippocampal and cerebellar cells. Int J Dev Neurosci 2009;27:233–41.

72. Darnaudery M, Perez-Martin M, Belizaire G, Maccari S, Garcia-Segura LM. Insulin-like growth factor 1 reduces age-related disorders induced by prenatal stress in female rats. Neurobiol Aging 2006;27:119–27. 73. Weinstock M. Gender differences in the effects of prenatal stress on brain development and behaviour. Neurochem Res 2007;32:1730–40. 74. Xu J, Deng X, Watkins R, Disteche CM. Sex-specific differences in expression of histone demethylases Utx and Uty in mouse brain and neurons. J Neurosci 2008;28:4521–7. 75. Carrel L, Willard HF. X-inactivation profile reveals extensive variability in X-linked gene expression in females. Nature 2005;434:400–4. 76. Smith-Bouvier DL, Divekar AA, Sasidhar M, Du S, TiwariWoodruff SK, King JK, Arnold AP, Singh RR, Voskuhl RR. A role for sex chromosome complement in the female bias in autoimmune disease. J Exp Med 2008;205:1099–108. 77. Melcangi RC, Panzica G. Neuroactive steroids: an update of their roles in central and peripheral nervous system. Psychoneuroendocrinology 2009;34:S1–8. 78. Schumacher M, Guennoun R, Stein DG, De Nicola AF. Progesterone: therapeutic opportunities for neuroprotection and myelin repair. Pharmacol Ther 2007;116:77–106. 79. Panzica GC, Melcangi RC. The endocrine nervous system: source and target for neuroactive steroids. Brain Res Rev 2008;57:271–6. 80. Garcia-Segura LM, Veiga S, Sierra A, Melcangi RC, Azcoitia I. Aromatase: a neuroprotective enzyme. Prog Neurobiol 2003;71:31–41. 81. Garcia-Segura LM, Melcangi RC. Steroids and glial cell function. Glia 2006;54:485–98. 82. Garcia-Segura LM, Balthazart J. Steroids and neuroprotection: new advances. Front Neuroendocrinol 2009;30:v–ix. 83. Melcangi RC, Garcia-Segura LM, Mensah-Nyagan AG. Neuroactive steroids: state of the art and new perspectives. Cell Mol Life Sci 2008;65:777–97. 84. Azcoitia I, Fernandez-Galaz C, Sierra A, Garcia-Segura LM. Gonadal hormones affect neuronal vulnerability to excitotoxininduced degeneration. J Neurocytol 1999;28:699–710. 85. Azcoitia I, Sierra A, Veiga S, Honda S, Harada N, GarciaSegura LM. Brain aromatase is neuroprotective. J Neurobiol 2001;47:318–29. 86. Hoffman GE, Merchenthaler I, Zup SL. Neuroprotection by ovarian hormones in animal models of neurological disease. Endocrine 2006;29:217–31. 87. Yang SH, Perez E, Cutright J, Liu R, He Z, Day AL, Simpkins JW. Testosterone increases neurotoxicity of glutamate in vitro and ischemia-reperfusion injury in an animal model. J Appl Physiol 2002;92:195–201. 88. Cheng J, Alkayed NJ, Hurn PD. Deleterious effects of dihydrotestosterone on cerebral ischemic injury. J Cereb Blood Flow Metab 2007;27:1553–62. 89. Hawk T, Zhang YQ, Rajakumar G, Day AL, Simpkins JW. Testosterone increases and estradiol decreases middle cerebral artery occlusion lesion size in male rats. Brain Res 1998; 796:296–8. 90. de Olmos S, Bueno A, Bender C, Lorenzo A, de Olmos J. Sex differences and influence of gonadal hormones on MK801induced neuronal degeneration in the granular retrosplenial cortex of the rat. Brain Struct Funct 2008;213:229–38. 91. Gillies GE, McArthur S. Independent influences of sex steroids of systemic and central origin in a rat model of Parkinson’s disease: a contribution to sex-specific neuroprotection by estrogens. Horm Behav 2010;57:23–34.

Article in press - uncorrected proof Melcangi and Garcia-Segura: Sex dimorphic features in the brain

92. Gillies GE, Murray HE, Dexter D, McArthur S. Sex dimorphisms in the neuroprotective effects of estrogen in an animal model of Parkinson’s disease. Pharmacol Biochem Behav 2004;78:513–22. 93. Morale MC, L’Episcopo F, Tirolo C, Giaquinta G, Caniglia S, Testa N, Arcieri P, Serra PA, Lupo G, Alberghina M, Harada N, Honda S, Panzica GC, Marchetti B. Loss of aromatase cytochrome P450 function as a risk factor for Parkinson’s disease? Brain Res Rev 2008;57:431–43. 94. McArthur S, Murray HE, Dhankot A, Dexter DT, Gillies GE. Striatal susceptibility to a dopaminergic neurotoxin is independent of sex hormone effects on cell survival and DAT expression but is exacerbated by central aromatase inhibition. J Neurochem 2007;100:678–92. 95. Yu L, Liao PC. Sexual differences and estrous cycle in methamphetamine-induced dopamine and serotonin depletions in the striatum of mice. J Neural Transm 2000;107:419–27. 96. Dluzen D, Horstink M. Estrogen as neuroprotectant of nigrostriatal dopaminergic system: laboratory and clinical studies. Endocrine 2003;21:67–75. 97. Datla KP, Murray HE, Pillai AV, Gillies GE, Dexter DT. Differences in dopaminergic neuroprotective effects of estrogen during estrous cycle. Neuroreport 2003;14:47–50. 98. Rodriguez-Navarro JA, Solano RM, Casarejos MJ, Gomez A, Perucho J, de Yebenes JG, Mena MA. Gender differences and estrogen effects in parkin null mice. J Neurochem 2008; 106:2143–57. 99. Meffre D, Pianos A, Liere P, Eychenne B, Cambourg A, Schumacher M, Stein DG, Guennoun R. Steroid profiling in brain and plasma of male and pseudopregnant female rats after traumatic brain injury: analysis by gas chromatography/mass spectrometry. Endocrinology 2007;148:2505–17. 100. di Michele F, Lekieffre D, Pasini A, Bernardi G, Benavides J, Romeo E. Increased neurosteroids synthesis after brain and spinal cord injury in rats. Neurosci Lett 2000;284:65–8. 101. Pesaresi M, Maschi O, Giatti S, Garcia-Segura LM, Caruso D, Melcangi RC. Sex differences in neuroactive steroid levels in the nervous system of diabetic and non-diabetic rats. Horm Behav 2010;57:46–55. 102. Giatti S, D’Intino G, Maschi O, Pesaresi M, Garcia-Segura LM, Calza L, Caruso D, Melcangi RC. Acute experimental autoimmune encephalomyelitis induces sex dimorphic changes in neuroactive steroid levels. Neurochem Int 2010;56: 118–27. 103. Caruso D, D’Intino G, Giatti S, Maschi O, Pesaresi M, Calabrese D, Garcia-Segura LM, Calza L, Melcangi RC. Sexdimorphic changes in neuroactive steroid levels after chronic experimental autoimmune encephalomyelitis. J Neurochem 2010;114:921–32. 104. Caruso D, Scurati S, Maschi O, De Angelis L, Roglio I, Giatti S, Garcia-Segura LM, Melcangi RC. Evaluation of neuroactive steroid levels by liquid chromatography-tandem mass spectrometry in central and peripheral nervous system: effect of diabetes. Neurochem Int 2008;52:560–8. 105. Ter Horst GJ, Wichmann R, Gerrits M, Westenbroek C, Lin Y. Sex differences in stress responses: focus on ovarian hormones. Physiol Behav 2009;97:239–49. 106. Galea LA. Gonadal hormone modulation of neurogenesis in the dentate gyrus of adult male and female rodents. Brain Res Rev 2008;57:332–41. 107. Swamydas M, Bessert D, Skoff R. Sexual dimorphism of oligodendrocytes is mediated by differential regulation of signaling pathways. J Neurosci Res 2009;87:3306–19.

391

108. Zhang L, Li B, Zhao W, Chang YH, Ma W, Dragan M, Barker JL, Hu Q, Rubinow DR. Sex-related differences in MAPKs activation in rat astrocytes: effects of estrogen on cell death. Brain Res Mol Brain Res 2002;103:1–11. 109. Westberry JM, Prewitt AK, Wilson ME. Epigenetic regulation of the estrogen receptor alpha promoter in the cerebral cortex following ischemia in male and female rats. Neuroscience 2008;152:982–9. 110. Fish EN. The X-files in immunity: sex-based differences predispose immune responses. Nat Rev Immunol 2008;8:737–44. 111. Barna M, Komatsu T, Bi Z, Reiss CS. Sex differences in susceptibility to viral infection of the central nervous system. J Neuroimmunol 1996;67:31–9. 112. Pelfrey CM, Cotleur AC, Lee JC, Rudick RA. Sex differences in cytokine responses to myelin peptides in multiple sclerosis. J Neuroimmunol 2002;130:211–23. 113. Moldovan IR, Cotleur AC, Zamor N, Butler RS, Pelfrey CM. Multiple sclerosis patients show sexual dimorphism in cytokine responses to myelin antigens. J Neuroimmunol 2008;193: 161–9. 114. Sinha S, Kaler LJ, Proctor TM, Teuscher C, Vandenbark AA, Offner H. IL-13-mediated gender difference in susceptibility to autoimmune encephalomyelitis. J Immunol 2008;180: 2679–85. 115. Reddy J, Waldner H, Zhang X, Illes Z, Wucherpfennig KW, Sobel RA, Kuchroo VK. Cutting edge: CD4qCD25q regulatory T cells contribute to gender differences in susceptibility to experimental autoimmune encephalomyelitis. J Immunol 2005;175:5591–5. 116. Matarese G, Sanna V, Di Giacomo A, Lord GM, Howard JK, Bloom SR, Lechler RI, Fontana S, Zappacosta S. Leptin potentiates experimental autoimmune encephalomyelitis in SJL female mice and confers susceptibility to males. Eur J Immunol 2001;31:1324–32. 117. Bebo BF, Jr., Adlard K, Schuster JC, Unsicker L, Vandenbark AA, Offner H. Gender differences in protection from EAE induced by oral tolerance with a peptide analogue of MBP-Ac1-11. J Neurosci Res 1999;55:432–40. 118. Pozzilli C, Tomassini V, Marinelli F, Paolillo A, Gasperini C, Bastianello S. ‘Gender gap’ in multiple sclerosis: magnetic resonance imaging evidence. Eur J Neurol 2003;10:95–7. 119. Gimenez-Llort L, Arranz L, Mate I, De la Fuente M. Genderspecific neuroimmunoendocrine aging in a triple-transgenic 3xTg-AD mouse model for Alzheimer’s disease and its relation with longevity. Neuroimmunomodulation 2008;15:331– 43. 120. Ciesielska A, Joniec I, Kurkowska-Jastrzebska I, Przybylkowski A, Gromadzka G, Czlonkowska A, Czlonkowski A. Influence of age and gender on cytokine expression in a murine model of Parkinson’s disease. Neuroimmunomodulation 2007;14:255–65. 121. Ballerio R, Gianazza E, Mussoni L, Miller I, Gelosa P, Guerrini U, Eberini I, Gemeiner M, Belcredito S, Tremoli E, Sironi L. Gender differences in endothelial function and inflammatory markers along the occurrence of pathological events in stroke-prone rats. Exp Mol Pathol 2007;82:33–41. 122. Arevalo MA, Santos-Galindo M, Bellini MJ, Azcoitia I, Garcia-Segura LM. Actions of estrogens on glial cells: implications for neuroprotection. Biochim Biophys Acta 2010; 1800:1106–12. 123. Butts CL, Sternberg EM. Neuroendocrine factors alter host defense by modulating immune function. Cell Immunol 2008;252:7–15.

Copyright of Hormone Molecular Biology & Clinical Investigation is the property of De Gruyter and its content may not be copied or emailed to multiple sites or posted to a listserv without the copyright holder's express written permission. However, users may print, download, or email articles for individual use.

Sex differences in the injured brain.

Observations obtained in human and in experimental models clearly demonstrate sex differences in degenerative events occurring in the central nervous ...
212KB Sizes 0 Downloads 7 Views