当前位置: 首页 > 医学版 > 期刊论文 > 内科学 > 内分泌进展 > 2005年 > 第3期 > 正文
编号:11168588
Are Estrogens Protective or Risk Factors in Brain Injury and Neurodegeneration? Reevaluation after the Women’s Health Initiative
http://www.100md.com 内分泌进展 2005年第3期
     Division of Biological Sciences, University of California Davis, Davis, California 95616-8536

    Abstract

    Estrogens are essential for normal reproductive function. In addition, they exert important, complex, and diverse nonreproductive actions on multiple tissues. Although accumulating evidence from basic science studies using animal models suggests that estradiol plays a critical neuroprotective role against multiple types of neurodegenerative diseases and injuries, recent clinical studies have reported either inconclusive or untoward effects of hormone therapy on the brain. We focus herein on the work that we have done during the past 6 yr that strongly suggests that low levels of estradiol therapy exert dramatic protective actions in the adult injured brain. Our results reveal that 17?-estradiol slows the progression of this injury and diminishes the extent of cell death by suppressing apoptotic cell death pathways and enhancing expression of genes that optimize cell survival. Furthermore, we have found that estrogen receptors play a pivotal functional role in neuroprotection. Together, these results carry broad implications for the selective targeting of estrogen receptors in the treatment of neurodegenerative conditions resulting from disease or injury, particularly for aging, postmenopausal women.

    I. Introduction

    II. Definition and Incidence of Cerebrovascular Stroke

    III. Experimental Models of Stroke and the Role of Estrogens

    IV. Summary

    I. Introduction

    NUMEROUS BASIC SCIENCE studies using animal models and in vitro cell and explant cultures, observational studies performed with human populations, and clinical trials led to the belief that ovarian hormones play an important role in providing women with protection against neurodegenerative diseases. However, whether hormone therapy (HT) in postmenopausal women is beneficial has become controversial because of the seemingly contradictory results when we compare these earlier studies with the recent results of the Women’s Health Initiative (WHI) (1, 2, 3, 4). The results that emerged from these clinical trials showed that use of conjugated equine estrogen (Premarin) with and without simultaneous medroxyprogesterone acetate (Prempro) lacked treatment benefit and increased risk of ischemic stroke. Before we accept these findings as the final word on the effects of HT on the adult brain, we should consider that this clinical trial used a specific HT regime on a group of older postmenopausal women, many of whom were obese. Whether or not different hormone preparations, initiated during the perimenopausal transition with little interruption with the normal menstrual cyclicity, would be efficacious remains unknown.

    In our studies, we have used an animal model of cerebrovascular stroke to assess whether estradiol influences the extent of injury, under what circumstances it protects, and the mechanisms that underlie protective actions. We should emphasize that to date, most clinical studies have assessed whether estradiol therapy (ET)/HT alter the risk and mortality of stroke, but studies have not addressed whether estradiol decreases the extent of brain injury resulting from stroke or the cellular and molecular basis of decreased injury or improved repair. Most basic science studies have focused attention on whether ET/HT decrease the extent of injury and cell death after a stroke event has been experimentally induced. This is a very different question and must be appreciated when we compare data from clinical vs. basic science studies.

    II. Definition and Incidence of Cerebrovascular Stroke

    Cerebrovascular stroke has been classified into two broad categories: 1) ischemic stroke, in which there is an interruption of blood flow to the brain due to an embolism that becomes lodged in a cerebral vessel; or 2) hemorrhagic stroke, which involves a bursting of cerebral vessels or compression of brain tissue. According to the American Heart Association, ischemic stroke is over four times more common than hemorrhagic stroke (see Ref. 5 and http://www.americanheart.org). In stroke, decreased cerebral blood flow deprives neurons and surrounding cells of crucial substrates (e.g., glucose and oxygen), and cells in the region will ultimately die. Neurons are particularly vulnerable to the ischemic insult. The pattern of cell death displayed depends upon the severity of the ischemic insult. Complete, permanent vessel occlusion leads to pannecrosis, in which all cell types in the affected area die due to metabolic failure. In areas where collateral vessels lead to maintenance of some, albeit decreased, blood flow, neurons will undergo a delayed form of cell death, or apoptosis. Therefore, in a typical ischemic stroke, there is often a core area of necrotic damage in brain regions supplied solely by the blocked vessel. A border zone, known as the ischemic penumbra, surrounds this core area. This area is less severely affected due to the presence of some level of collateral circulation supplied by branches of other major vessels. It is salvageable if the environment is optimized through administration of growth factors (6, 7, 8) or hormones (9, 10) or suppression of the inflammatory response (11).

    Ischemic stroke can involve a permanent blockage of a cerebral vessel by an embolism or a transient blockade of a cerebral vessel. Both the occlusion and the restoration of blood flow (reperfusion) to ischemic tissue cause cellular damage, possibly by different molecular mechanisms (12, 13, 14). Reperfusion injury may be mediated by various second messenger signaling pathways, particularly protein kinase C, unregulated neurotransmitter release, decrease in cellular energy stores, inhibition of electron transport, and inflammation (for review, see Ref. 15).

    The risk of stroke is lower in premenopausal women than in age-matched men and increases in postmenopausal women to become equal to that of men by the time women are over 65 yr old (see Ref. 16 and http://www.americanheart.org). This observation led investigators to postulate that ET and HT may protect postmenopausal women. Indeed, several studies over the past 20 yr supported this contention; however, the clinical and epidemiological studies were not uniformly positive. Paganini-Hill (17) reviewed the studies that were performed before 1995 and concluded that the bulk of the evidence suggests that ET decreased the risk of stroke. In 2001, the Women’s Estrogen for Stroke Trial indicated that estrogens do not protect against the rate of either nonfatal stroke or death in postmenopausal women with a history of stroke (18). The most recent findings of the WHI demonstrate that combined conjugated equine estrogens with and without concomitant medroxyprogesterone acetate exacerbates the risk of ischemic stroke in women who were postmenopausal for approximately 12 yr before the initiation of ET/HT (4, 19). The results of these studies are consistent with those that fail to detect estrogen-mediated protection of the heart in women who already suffer from cardiovascular disease (20) or of the brain in women who already exhibit signs of Alzheimer’s disease (21). Together, these studies suggest that estrogen does not effectively protect against or reverse a disease process that has already been initiated. They suggest that this hormone may be able to protect against the initial stages of disease progression but is not an effective hormone of repair when damage from disease has progressed to a significant degree.

    III. Experimental Models of Stroke and the Role of Estrogens

    We have used an animal model in which the middle cerebral artery is permanently occluded to examine the effects of estradiol on neurodegeneration (22). In all of our studies that we describe below, we have used exclusively the 17? form of estradiol. As discussed below, many studies, including our own, have investigated whether estradiol can attenuate cell death resulting from ischemic injury and whether the mechanisms of protection against cell death involve suppression of apoptotic signaling. We have found that low, physiological doses of ET are sufficient to exert dramatic protection against stroke injury in young female rats (Fig. 1) (23). Our endocrine paradigm produces fairly steady levels of approximately 15 pg/ml of estradiol in serum, which are strikingly similar to the levels we observe in rats during all days of the estrous cycle, except on proestrus, when the rise is in preparation for the ovulatory surge of gonadotropins. Using these concentrations of estradiol, we found that ET must be administered before middle cerebral artery occlusion and that treatment at the time of the injury does not decrease the extent of the infarct. We found that the protection does not involve estradiol-enhanced blood flow or increased blood glucose levels. In addition, we found that ET did not protect against the initial stages of injury, but instead protected against the later stages of injury. These initial results clearly established that very low levels of estradiol exert profound neuroprotective actions against stroke-like injury. They implied that ET protected predominantly by decreasing delayed cell death. Other researchers have used higher concentrations of estradiol, which reach 100 to 10,000 times the concentrations that we have used and found that animals can be treated up to 3 h after middle cerebral artery occlusion and still benefit from exposure to estradiol and estrogen-like compounds (24). Thus, these higher concentrations of estrogens appear to act through very different mechanisms: 1) estrogens affect cerebral blood flow and may prevent injury by causing vasodilation; 2) estrogens increase blood glucose concentrations and may allow neurons to be in a more protective metabolic environment; 3) estrogens act as antioxidants and may prevent free radical generation-induced cell death; and 4) estrogens influence the production of nitric oxide and may influence vascular endothelial cell function to attenuate damage to the vasculature.

    We investigated whether estradiol-induced neuroprotection requires estrogen receptor (ER) and/or ER? and discovered that ER plays an essential functional role in protecting against cell death. Our data reveal that physiological levels of estradiol induce the reappearance of ER gene and protein expression on the ipsilateral side of injury (25). This gene is expressed at high levels during neonatal development when the cortex undergoes neurite outgrowth and synaptogenesis but decreases during later stages of development and in adulthood. Injury-induced reappearance of ER gene and protein expression may indicate that the injured brain provides signals that communicate the need for expression of a receptor that is required for reinitiation of "developmental" events in the injured cortex. We found that the presence of this ER subtype is essential to protect against cerebral ischemia (26, 27). We used ER or ER? knockout mice and found that when the ER is absent, estradiol no longer protected against middle cerebral artery occlusion-induced cell death (Fig. 2). In marked contrast, in the absence of ER?, estradiol continued to exert its protective effects. These studies clearly establish that the classic estrogen receptor, ER, which was once thought to be more important in reproductive functions than in nonreproductive functions, is the critical mechanistic link in the ability of low levels of estradiol to protect against delayed cell death.

    We have begun to assess the repertoire of downstream genomic targets of estradiol action through ERs. To date, we have reported that estradiol modulates the expression of multiple genes in ischemic brain injury, including ones that regulate the balance of cell death and cell survival factors (25) and immediate early genes, such as c-fos (28). Two examples of genes that respond to estradiol after middle cerebral artery occlusion illustrate the principle that multiple genes are either up-regulated or down-regulated.

    As a first example, Bcl-2 is a protooncogene that promotes cell survival in a variety of tissues including the brain. Because estradiol is known to promote cell survival via Bcl-2 in nonneural tissues, we tested the hypothesis that estradiol decreases cell death by influencing bcl-2 expression in ischemic brain injury. We analyzed gene expression in microdissected regions of the cerebral cortex adjacent to the infarct in ovariectomized rats and the equivalent regions of the brain in ET rats. Tissue samples were obtained from injured and sham female rats. We found that estradiol prevented the injury-induced down-regulation of bcl-2 expression on the ipsilateral side of the brain. This effect was specific to bcl-2, because expression of other members of the bcl-2 family (bax, bcl-xL, bcl-xS, bad, and bim) was unaffected by ET (Fig. 3).

    In the second example, immediate early genes are induced by various forms of brain injury, and their induction is known to be a critical step in apoptotic cell death. We tested the hypothesis that the ability of estradiol to protect the brain against middle cerebral artery occlusion-induced cell death involves attenuation of the expression of one or more immediate early genes. We assessed immediate early gene mRNAs in regions ipsilateral and contralateral to the region of injury. Our results reveal that c-fos, fosB, c-jun, and junB mRNA levels were up-regulated 24 h after middle cerebral artery occlusion. Furthermore, among these, estradiol affected only the expression of c-fos mRNA and protein expression on both sides of the brain: the steroid treatment attenuated the injury-induced increase in a time-specific manner (Fig. 4). Our findings strongly suggest that the ability of estradiol to protect the brain against the late stages of cell death involve the attenuation of c-fos induction.

    It is intriguing that injury influences some of these genes (bcl-2) only on the side of the infarct; whereas, in other cases, both sides of the brain are affected (c-fos). We interpret this to mean that even the uninjured side of the brain is involved in some aspects of protection and repair. Together, the up-regulation and down-regulation of gene expression enhance neuronal survival and suppress the expression of those that promote cell death.

    It is important to remember that even in animal models, estrogens do not always exert beneficial effects. Under certain circumstances, 17?-estradiol either fails to protect or even harms the brain. Although estradiol can decrease brain injury in the vast majority of studies, estradiol fails to attenuate cell death in some animal models (29, 30, 31, 32). Several points should be emphasized. First, although a large body of literature suggests that estrogens are potent neuroprotective factors under numerous experimental circumstances in vivo and in many in vitro experimental conditions, in other experimental and disease/injury paradigms, estrogens either fail to protect or even exacerbate neural injury. It is possible that when the degree of injury is too severe, as may be the case in the hippocampus after prolonged global ischemia or when a disease state is already in progress, the actions of estradiol cannot overcome this degree of injury and are not sufficient to prevent cell death. Under other circumstances, estrogens can be deleterious to neural function. For example, in animal models of epilepsy, estradiol lowers the threshold for seizures and facilitates the induction and duration of excitatory neural firing. These data suggest that estrogen therapy may not always exert only beneficial actions in the brains of postmenopausal women, particularly in those with a medical history of epilepsy. As we continue to learn about the complexity of estrogen action with regard to dose, type of estrogen, and neurological condition, we may be better able to modify and transform ET into therapies that exert beneficial effects without the negative actions. Indeed, if we were able to develop estrogen-like compounds that did not have feminizing effects, these compounds could be used in both men and women.

    IV. Summary

    In summary, a large number of clinical and basic science studies have led to a new appreciation that estradiol acts far beyond the reproductive axis and exerts profound protective actions in the adult and aging brain. Although we have only begun to identify potential cellular and molecular mechanisms of this protection, our growing knowledge of estrogen action in the injured brain will ultimately lead to a more complete understanding of the precise mechanisms underlying estradiol-mediated protection. This knowledge is crucial to developing both preventative and acute therapies for neurodegenerative conditions and carries great promise in improving the quality of life in our aging population.

    Footnotes

    This work was supported by the National Institutes of Health (Grants AG02224 and AG17164) and the Ellison Foundation.

    Current address for D.B.D.: Department of Neurology, University of California San Francisco, San Francisco, California 94143.

    Current address for S.W.R.: Department of Psychiatry, University of Virginia, Charlottesville, Virginia 22903.

    First Published Online April 19, 2005

    Abbreviations: ER, Estrogen receptor; ET, estradiol therapy; HT, hormone therapy.

    References

    Rossouw JE, Anderson GL, Prentice RL, LaCroix AZ, Kooperberg C, Stefanick ML, Jackson RD, Beresford SAA, Howard BV, Johnson KC, Kotchen JM, Ockene J 2002 Risks and benefits of estrogen plus progestin in healthy postmenopausal women: principal results from the Women’s Health Initiative randomized controlled trial. JAMA 288:321–333

    Rapp SR, Espeland MA, Shumaker SA, Henderson VW, Brunner RL, Manson JE, Gass MLS, Stefanick ML, Lane DS, Hays J, Johnson KC, Coker LH, Dailey M, Bowen D 2003 Effect of estrogen plus progestin on global cognitive function in postmenopausal women. JAMA 289:2663–2672

    Shumaker SA, Legault C, Rapp SR, Thal LJ, Wallace RB, Ockene JK, Hendrix SL, Jones BN, Annlouise RA, Jackson RD, Kotchen JM, Wassertheil-Smoller S, Wactawski-Wende J 2003 Estrogen plus progestin and the incidence of dementia and mild cognitive impairment in postmenopausal women. JAMA 289:2651–2662

    Anderson GL, Limacher M, Assaf AR, Bassford T, Beresford SA, Black H, Bonds D, Brunner R 2004 Effects of conjugated equine estrogen in postmenopausal women with hysterectomy: the Women’s Health Initiative randomized controlled trial. JAMA 291:1701–1712

    2002 Heart and stroke statistical update, 2001. Dallas, TX: American Heart Association

    Benraiss A, Chmielnicki E, Lerner K, Roh D, Goldman SA 2001 Adenoviral BDNF induces both neostriatal and olfactory neuronal recruitment from endogenous progenitor cells in the adult forebrain. J Neurosci 21:6718–6731

    Gustafson E, Andsberg G, Darsalia V, Mohapel P, Mandel RJ, Kirik D, Lindvall O, Kokaia Z 2003 Anterograde delivery of BDNF to striatum via nigral transduction of recombinant adeno-associated virus increases neuronal death but promotes neurogenic response following stroke. Eur J Neurosci 17:2667–2678

    Yoshimura S, Takagi Y, Harada J, Teramoto T, Thomas SS, Waeber C, Bakowska J, Breakefield XO, Moskowitz MA 2001 FGF-2 regulation of neurogenesis in adult hippocampus after brain injury. Proc Natl Acad Sci USA 98:5874–5879

    Ormerod BK, Lee TT-Y, Galea LAM 2003 Estradiol initially enhances but subsequently suppresses (via adrenal steroids) granule cell proliferation in the dentate gyrus of adult female rats. J Neurobiol 55:247–260

    Tanapat P, Hastings NB, Reeves AJ, Gould E 1999 Estrogen stimulates a transient increase in the number of new neurons in the dentate gyrus of the adult female rat. J Neurosci 19:5792–5801

    Monje ML, Toda H, Palmer TD 2003 Inflammatory blockade restores adult hippocampal neurogenesis. Science 302:1760–1765

    DeGracia DJ, Kumar R, Owen CR, Krause GS, White BC 2002 Molecular pathways of protein synthesis inhibition during brain reperfusion: implications for neuronal survival or death. J Cereb Blood Flow Metab 22:127–141

    Jean WC, Spellman BA, Nussbaum ES, Low WC 1998 Reperfusion injury after focal cerebral ischemia: the role of inflammation and the therapeutic horizon. Neurosurgery 43:1382–1397

    Bright R, Raval AP, Dembner JM, Perez-Pinzon MA, Steinberg GK, Yenari MA, Mochly-Rosen D 2004 Protein kinase C mediates cerebral reperfusion injury. Neurobiol Dis 24:6880–6888

    Lipton P 1999 Ischemic cell death in brain neurons. Physiol Rev 79:1431–1568

    2001 Heart and stroke statistical update, 2001. Dallas, TX: American Heart Association

    Paganini-Hill A 1995 Estrogen replacement therapy and stroke. Prog Cardiovasc Dis 38:223–242

    Viscoli CM, Brass LM, Kernan WN, Sarrel PM, Suissa S, Horwitz RI 2001 A clinical trial of estrogen-replacement therapy after ischemic stroke. N Engl J Med 345:1243–1249

    Wassertheil-Smoller S, Hendrix SL, Limacher M, Heiss G, Kooperberg C, Baird A, Kotchen T, Curb JD, Black H, Rossouw JE, Aragaki A, Safford M, Stein E, Laowattana S, Mysiw WJ 2003 Effect of estrogen plus progestin on stroke in postmenopausal women. JAMA 289:2673–2684

    Hulley S, Grady D, Bush T, Furberg C, Herrington D, Riggs B, Vittinghoff E 1998 Randomized trial of estrogen plus progestin for secondary prevention of coronary heart disease in postmenopausal women. JAMA 280:605–613

    Mulnard RA, Cotman CW, Kawas C, van Dyck CH, Sano M, Doody R, Koss E, Pfeiffer E, Jin S, Garnst A, Grundman M, Thomas R, Thal LJ 2000 Estrogen replacement therapy for treatment of mild to moderate Alzheimer’s disease. JAMA 283:1007–1015

    Longa EZ, Weinstein PR, Carlson S, Cummins R 1989 Reversible middle cerebral artery occlusion without craniectomy in rats. Stroke 20:84–91

    Dubal DB, Kashon ML, Pettigrew LC, Ren JM, Finklestein SP, Rau SW, Wise PM 1998 Estradiol protects against ischemic injury. J Cereb Blood Flow Metab 18:1253–1258

    Yang S-H, Shi J, Day AL, Simpkins JW 2000 Estradiol exerts neuroprotective effects when administered after ischemic insult. Stroke 31:745–750

    Dubal DB, Shughrue PJ, Wilson ME, Merchenthaler I, Wise PM 1999 Estradiol modulates Bcl-2 in cerebral ischemia: a potential role for estrogen receptors. J Neurosci 19:6385–6393

    Dubal DB, Zhu B, Yu B, Rau SW, Shughrue PJ, Merchenthaler I, Kindy MS, Wise PM 2001 Estrogen receptor-, not -?, is a critical link in estradiol-mediated protection against brain injury. Proc Natl Acad Sci USA 98:1952–1957

    Wilson ME, Dubal DB, Wise PM 2000 Estradiol protects against injury-induced cell death in cortical explant cultures: a role for estrogen receptors. Brain Res 873:235–242

    Rau SW, Dubal DB, B?ttner M, Wise PM 2003 Estradiol differentially regulates c-fos after focal cerebral ischemia. J Neurosci 23:10487–10494

    Santizo RA, Xu HL, Ye S, Baughman VL, Pelligrino DA 2001 Loss of benefit from estrogen replacement therapy in diabetic ovariectomized female rats subjected to transient forebrain ischemia. Brain Res 956:86–95

    Harukuni I, Hurn PD, Crain BJ 2001 Deleterious effect of ?-estradiol in a rat model of transient forebrain ischemia. Brain Res 900:137–142

    Emerson CS, Headrick JP, Vink R 1993 Estrogen improves biochemical and neurologic outcome following traumatic brain injury in male rats, but not in females. Brain Res 608:95–100

    Kondo Y, Suzuki K, Sakuma Y 1997 Estrogen alleviates cognitive dysfunction following transient brain ischemia in ovariectomized gerbils. Neurosci Lett 238:45–48(Phyllis M. Wise, Dena B. )