351
|
Koerner IP, Zhang W, Cheng J, Parker S, Hurn PD, Alkayed NJ. Soluble epoxide hydrolase: regulation by estrogen and role in the inflammatory response to cerebral ischemia. FRONTIERS IN BIOSCIENCE : A JOURNAL AND VIRTUAL LIBRARY 2008; 13:2833-41. [PMID: 17981757 PMCID: PMC2593891 DOI: 10.2741/2889] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The protection from ischemic brain injury enjoyed by females is linked to the female sex hormone 17beta-estradiol. We tested the hypothesis that neuroprotection by estradiol entails the prevention of ischemia-induced inflammatory response, through suppression of the P450 eicosanoids-metabolizing enzyme soluble epoxide hydrolase (sEH). Ovariectomized female rats with and without estradiol replacement underwent 2-hour middle cerebral artery occlusion (MCAO). SEH expression was determined using Western blot, and inflammatory cytokine mRNA levels were measured at 6, 24 and 48 hours after MCAO. Cytokine mRNA was also measured in sEH-knockout mice, and in rats treated with sEH inhibitors. Estradiol reduced basal and post-ischemic sEH expression. MCAO strongly induced mRNA levels of tumor necrosis factor-alpha, interleukin 6, and interleukin 1beta, which was attenuated in sEH-knockouts, but not by sEH inhibitors. Estradiol replacement exhibited a bimodal effect on cytokine mRNA, with increased early and reduced delayed expression. While estradiol suppresses cerebral sEH expression, and sEH suppression diminishes inflammation after MCAO, our findings suggest that the effect of estrogen on inflammation is complex, and only partially explained by sEH suppression.
Collapse
Affiliation(s)
- Ines P Koerner
- Department of Anesthesiology and Peri-Operative Medicine, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | | | | | | | | | | |
Collapse
|
352
|
|
353
|
Singh M, Sumien N, Kyser C, Simpkins JW. Estrogens and progesterone as neuroprotectants: what animal models teach us. FRONTIERS IN BIOSCIENCE : A JOURNAL AND VIRTUAL LIBRARY 2008; 13:1083-9. [PMID: 17981614 PMCID: PMC2586167 DOI: 10.2741/2746] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Estradiol and progesterone are two steroid hormones that target a variety of organ systems, including the heart, the bone and the brain. With respect to the latter, a large volume of basic science studies support the neuroprotective role of estradiol and/or progesterone. In fact, the results of such studies prompted the assessment of these hormones as protective agents against such disorders as Alzheimer's disease, stroke and traumatic brain injury. Interestingly, results from the Women's Health Initiative (WHI) yielded results that appeared to be inconsistent with the data derived from in vitro and in vivo models. However, we argue that the results from the basic science studies were not inconsistent with the clinical trials, but rather, are consistent with, and may even have predicted, the results from the WHI. To illustrate this point, we review here certain in vivo paradigms that have been used to assess the protective effects of estrogens and progesterone, and describe how the results from these animal models point to the importance of the type of hormone, the age of the subjects and the method of hormone administration, in determining whether or not hormones are neuroprotective.
Collapse
Affiliation(s)
- Meharvan Singh
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, USA.
| | | | | | | |
Collapse
|
354
|
Abstract
The human costs of stroke are very large and growing; it is the third largest cause of death in the United States and survivors are often faced with loss of ability to function independently. There is a large need for therapeutic approaches that act to protect neurons from the injury produced by ischemia and reperfusion. The goal of this review is to introduce and discuss the available data that endogenous cannabinoid signaling is altered during ischemia and that it contributes to the consequences of ischemia-induced injury. Overall, the available data suggest that inhibition of CB1 receptor activation together with increased CB2 receptor activation produces beneficial effects.
Collapse
Affiliation(s)
- Cecilia J Hillard
- Department of Pharmacology and Toxicology, Medical College of Wisconsin; Milwaukee, WI 53226, USA.
| |
Collapse
|
355
|
McCullough LD, Koerner IP, Hurn PD. Effects of gender and sex steroids on ischemic injury. HANDBOOK OF CLINICAL NEUROLOGY 2008; 92:149-69. [PMID: 18790274 DOI: 10.1016/s0072-9752(08)01908-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
356
|
Olsen TS, Dehlendorff C, Andersen KK. Sex-related time-dependent variations in post-stroke survival--evidence of a female stroke survival advantage. Neuroepidemiology 2007; 29:218-25. [PMID: 18075278 DOI: 10.1159/000112464] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Women live longer than men, yet most studies show that gender has no influence on survival after stroke. METHODS A registry was started in 2001, with the aim of registering all hospitalized stroke patients in Denmark, and it now holds 39,484 patients of which 48% are female. We studied the influence of gender on post-stroke mortality, from the time of admission through the subsequent years until death or censoring (mean follow-up time: 538 days). All patients underwent an evaluation including stroke severity, computed tomography and cardiovascular risk factors. Independent predictors of death were identified by means of a survival model based on 22,222 individuals with a complete data set. RESULTS Females were older and had severer stroke. Interestingly, the risk of death between genders was time dependent. The female/male stroke mortality rate favoured women from the first day of stroke and remained so during the first month suggesting a female survival advantage. Throughout the second month the rate reversed in favour of men suggesting that women in that period are paying a 'toll' for their initial survival advantage. Hereafter, the rate steadily decreased, and after 4 months women continued to have the same low risk as in the first week. CONCLUSIONS Our study suggests a female superiority in stroke survival competence.
Collapse
|
357
|
Saleh TM, Cribb AE, Connell BJ. Role of estrogen in central nuclei mediating stroke-induced changes in autonomic tone. J Stroke Cerebrovasc Dis 2007; 12:182-95. [PMID: 17903925 DOI: 10.1016/s1052-3057(03)00080-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2002] [Revised: 06/09/2003] [Accepted: 06/13/2003] [Indexed: 11/21/2022] Open
Abstract
The current investigation examined the role of estrogen in central autonomic regulatory nuclei on the autonomic dysfunction resulting from middle cerebral artery occlusion (MCAO). Experiments were done in anaesthetized male Sprague-Dawley rats. The effect of MCAO on autonomic tone was assessed by monitoring vagal and renal efferent nerve activities before and following systemic administration of either estrogen or saline and the bilateral microinjection of the estrogen receptor antagonist, ICI 182, 780, into several autonomic nuclei (the intrathecal space of the spinal cord, nucleus tractus solitarius, nucleus ambiguus, rostral ventrolateral medulla, parabrachial nucleus, central nucleus of the amygdala or ventral posteromedial thalamus). Autonomic reflex function was evoked using intravenous injection of increasing doses of phenylephrine (0.025-0.1 mg/kg) and the peak changes in heart rate and blood pressure were plotted to obtain the baroreflex sensitivity. The presence of ICI 182, 780 in the intrathecal space of the spinal cord, nucleus ambiguous, nucleus tractus solitarius, rostral ventrolateral medulla, parabrachial nucleus, or central nucleus of the amygdala prior to the administration of estrogen resulted in a significant attenuation (ranging from 79% to 94 %) in the estrogen-induced recovery of autonomic function following MCAO. Blocking estrogen receptors in the ventral posteromedial thalamus had no effect on the ability of estrogen to prevent the MCAO-induced changes in autonomic function. These results suggest that the estrogen-mediated recovery of autonomic function following MCAO is dependent on the availability of estrogen receptors in several forebrain and brainstem autonomic nuclei.
Collapse
Affiliation(s)
- Tarek M Saleh
- Department of Biomedical Sciences, Atlantic Veterinary College, University of Prince Edward Island, Charlottetown, Prince Edward Island, Canada.
| | | | | |
Collapse
|
358
|
Saleh TM, Connell BJ, Legge C, Cribb AE. Stroke-induced changes in estrogen release and neuronal activity in the parabrachial nucleus of the male rat. J Stroke Cerebrovasc Dis 2007; 13:24-34. [PMID: 17903946 DOI: 10.1016/j.jstrokecerebrovasdis.2004.01.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2003] [Accepted: 01/03/2004] [Indexed: 11/23/2022] Open
Abstract
Recent investigations have provided evidence to suggest exogenous estrogen administration into autonomic nuclei prevents or reverses the autonomic dysfunction observed after middle cerebral artery occlusion (MCAO) in male rats. Because estrogen seems to be a potent neuroprotectant against autonomic dysfunction, it is our hypothesis that endogenous estrogen levels within autonomic nuclei will increase in response to stroke. Therefore, in this investigation, in vivo microdialysis was used to simultaneously measure the concentration of estrogen in the plasma and in the parabrachial nucleus (PBN) of male Sprague-Dawley rats after MCAO. Analysis of dialysate samples before MCAO and in sham-operated controls revealed a baseline concentration of estrogen in the PBN (38 +/- 3 pg/mL; n = 36), which was significantly greater than that found in plasma (22 +/- 6 pg/mL; n = 6; P < .05). The concentration of estrogen in the PBN was significantly increased immediately after MCAO (85 +/- 4 pg/mL; n = 7; P < .05) but then decreased to below pre-MCAO values (12 +/- 2 pg/mL; n = 7; P < .05) by 90 minutes after MCAO and remained below baseline levels until the end of the experiment (240 minutes post-MCAO). No changes in plasma estrogen levels were detected at any time point after MCAO. In addition, extracellular electrophysiological recordings from PBN neurons revealed that MCAO resulted in an immediate decrease in the activity of PBN neurons, which was completely blocked after systemic estrogen injection. These results suggest that estrogen is released into the PBN in response to MCAO and that the source of estrogen seems to be primarily caused by terminal release as opposed to increased local synthesis.
Collapse
Affiliation(s)
- Tarek M Saleh
- Department of Biomedical Sciences, Atlantic Veterinary College, Charlottetown, Prince Edward Island, Canada.
| | | | | | | |
Collapse
|
359
|
Renolleau S, Fau S, Charriaut-Marlangue C. Gender-related differences in apoptotic pathways after neonatal cerebral ischemia. Neuroscientist 2007; 14:46-52. [PMID: 17971506 DOI: 10.1177/1073858407308889] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Many central nervous system (CNS) diseases display sexual dimorphism, specifically a predilection for one gender or a gender-dependent response to treatment. Exposure to circulating sex steroids is felt to be a chief contributor to this phenomenon. However, CNS diseases of childhood and of the elderly also demonstrate gender predominance and/or sexual dimorphism response to therapies. In this short update, we provide information concerning one of the most interesting new emerging concepts related to the influence of the sex in the pathogenesis of developmental brain injuries leading to different levels of neuroprotection between genders after cerebral hypoxia-ischemia or ischemia.
Collapse
|
360
|
Rigsby CS, Burch AE, Ogbi S, Pollock DM, Dorrance AM. Intact female stroke-prone hypertensive rats lack responsiveness to mineralocorticoid receptor antagonists. Am J Physiol Regul Integr Comp Physiol 2007; 293:R1754-63. [PMID: 17670862 PMCID: PMC2804103 DOI: 10.1152/ajpregu.00145.2007] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Data from the Framingham Heart Study suggest that women may be more sensitive to the deleterious cardiovascular remodeling effects of aldosterone. Previous studies from our laboratory have shown that chronic treatment with spironolactone, a mineralocorticoid receptor (MR) antagonist, decreases ischemic cerebral infarct size and prevents remodeling of the middle cerebral artery (MCA) in male spontaneously hypertensive stroke-prone rats (SHRSP). Therefore, we hypothesized that MR antagonism would reduce ischemic infarct size and prevent MCA remodeling in female SHRSP. Six-week-old female SHRSP were treated for 6 wk with spironolactone (25 or 50 mg.kg(-1).day(-1)) or eplerenone (100 mg.kg(-1).day(-1)) and compared with untreated controls. At 12 wk, cerebral ischemia was induced for 18 h using the intraluminal suture occlusion technique, or the MCA was isolated for analysis of passive structure using a pressurized arteriograph. MR antagonism had no effect on infarct size or passive MCA structure in female SHRSP. To study the potential effects of estrogen, the above experiments were repeated in bilaterally ovariectomized (OVX) female SHRSP treated with spironolactone (25 mg.kg(-1).day(-1)). Infarct size and vessel structure in OVX SHRSP were not different from control SHRSP. Spironolactone had no effect on infarct size in OVX SHRSP. However, MCA lumen and outer diameters were increased in spironolactone-treated OVX SHRSP, suggesting an effect of estrogen. Cerebral artery MR expression, assessed by Western blotting, was increased in female, compared with male, SHRSP. These studies highlight an apparent sexual dimorphism of MR expression and activity in the cerebral vasculature from hypertensive rats.
Collapse
Affiliation(s)
- Christiné S Rigsby
- Dept. of Physiology, Medical College of Georgia, 1120 15th St. (CA2091) Augusta, Georgia 30912-3000, USA.
| | | | | | | | | |
Collapse
|
361
|
Abstract
1. In recent years, the role of oestrogen in women's health has been a subject of considerable scientific and popular debate. There is unquestionable evidence that oestrogen has both potent and long-lasting effects on several vital organ systems, including the cardiovascular system, the autonomic nervous system and, most recently, within the central nervous system itself. 2. The research and medical community continues to debate whether the benefits of oestrogen therapy outweigh the risks in the treatment of the symptoms of menopause, the attenuation of the risk for cardiovascular insults, such as stroke and heart disease, and even the retardation of the progression of Alzheimer's disease. 3. The recent evidence provided by the Heart and Estrogen/Progestin Replacement Study (HERS) II clinical trial suggesting that long-term exposure to combined oestrogen and progestin in post-menopausal women who have previously had a heart attack or stroke (for secondary prevention) may actually increase their risk of a subsequent cardiovascular insult has further fuelled the debate. However, there remain considerable gaps in our knowledge with respect to the actual mechanisms by which oestrogen exerts its various beneficial effects at the cellular level for the primary prevention of cardiovascular disease. This information is essential if we are to harness the positive aspects of oestrogen therapy in such a manner as to avoid or minimize the associated risks of increased oestrogen exposure in women who we know, with some certainty, to be at an increased risk of cancers of the uterus, cervix and breast tissue.
Collapse
Affiliation(s)
- T M Saleh
- Department of Biomedical Science, Atlantic Veterinary College and The Prince Edward Island Health Research Institute, University of Prince Edward Island, Charlottetown, Prince Edward Island, Canada.
| | | |
Collapse
|
362
|
Abstract
Outcome from cerebral ischemia is sexually dimorphic in many experimental models. Male animals display greater sensitivity to ischemic injury than do their female counterparts; however, the underlying mechanism is unclear. The present study determined if the potent and nonaromatizable androgen, dihydrotestosterone (DHT), exacerbates ischemic damage in the male rat and alters postischemic gene expression after middle cerebral artery occlusion. At 22 h reperfusion, removal of androgens by castration provided protection from ischemic injury in both cortex and striatum (2,3,5-triphenyltetrazolium chloride (TTC) histology), whereas DHT replacement (50 mg subcutaneous implant) restored infarction volume to that of the intact male; testosterone (50 mg) had similar but less potent effects. We utilized microarray and real-time quantitative polymerase chain reaction (PCR) to identify genes differentially expressed at 6 h reperfusion in periinfarct cortex from castrated rats with or without DHT replacement. We identify, for the first time, a number of gene candidates that are induced by DHT with or without ischemia, many of which could account for cell death through enhanced inflammation, dysregulation of blood-brain barrier and the extracellular matrix, apoptosis, and ionic imbalance. Our data suggest that androgens are important mediators of ischemic damage in male brain and that transcriptional mechanisms should be considered as we seek to understand innate male sensitivity to cerebral ischemia.
Collapse
Affiliation(s)
- Jian Cheng
- Department of Anesthesiology & Peri-Operative Medicine, Oregon Health & Science University, Portland, Oregon 97239-3098, USA
| | | | | |
Collapse
|
363
|
Amantea D, Spagnuolo P, Bari M, Fezza F, Mazzei C, Tassorelli C, Morrone LA, Corasaniti MT, Maccarrone M, Bagetta G. Modulation of the endocannabinoid system by focal brain ischemia in the rat is involved in neuroprotection afforded by 17beta-estradiol. FEBS J 2007; 274:4464-775. [PMID: 17666109 DOI: 10.1111/j.1742-4658.2007.05975.x] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Endogenous levels of the endocannabinoid anandamide, and the activities of the synthesizing and hydrolyzing enzymes, i.e. N-acylphosphatidylethanolamine-hydrolyzing phospholipase D and fatty acid amide hydrolase, respectively, were determined in the cortex and the striatum of rats subjected to transient middle cerebral artery occlusion. Anandamide content was markedly increased ( approximately 3-fold over controls; P < 0.01) in the ischemic striatum after 2 h of middle cerebral artery occlusion, but not in the cortex, and this elevation was paralleled by increased activity of N-acylphosphatidylethanolamine-hydrolyzing phospholipase D ( approximately 1.7-fold; P < 0.01), and reduced activity ( approximately 0.6-fold; P < 0.01) and expression ( approximately 0.7-fold; P < 0.05) of fatty acid amide hydrolase. These effects of middle cerebral artery occlusion were further potentiated by 1 h of reperfusion, whereas anandamide binding to type 1 cannabinoid and type 1 vanilloid receptors was not affected significantly by the ischemic insult. Additionally, the cannabinoid type 1 receptor antagonist SR141716, but not the receptor agonist R-(+)-WIN55,212-2, significantly reduced (33%; P < 0.05) cerebral infarct volume detected 22 h after the beginning of reperfusion. A neuroprotective intraperitoneal dose of 17beta-estradiol (0.20 mg x kg(-1)) that reduced infarct size by 43% also minimized the effect of brain ischemia on the endocannabinoid system, in an estrogen receptor-dependent manner. In conclusion, we show that the endocannabinoid system is implicated in the pathophysiology of transient middle cerebral artery occlusion-induced brain damage, and that neuroprotection afforded by estrogen is coincident with a re-establishment of anandamide levels in the ischemic striatum through a mechanism that needs to be investigated further.
Collapse
Affiliation(s)
- Diana Amantea
- Department of Pharmacobiology and University Center for the Study of Adaptive Disorder and Headache (UCADH), Section of Neuropharmacology of Normal and Pathological Neuronal Plasticity, University of Calabria, Rende (CS), Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
364
|
Dziennis S, Jia T, Rønnekleiv OK, Hurn PD, Alkayed NJ. Role of signal transducer and activator of transcription-3 in estradiol-mediated neuroprotection. J Neurosci 2007; 27:7268-74. [PMID: 17611279 PMCID: PMC2570353 DOI: 10.1523/jneurosci.1558-07.2007] [Citation(s) in RCA: 96] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2007] [Revised: 05/19/2007] [Accepted: 05/28/2007] [Indexed: 01/03/2023] Open
Abstract
Estradiol is protective in experimental cerebral ischemia, but the precise mechanisms remain unknown. Signal transducer and activator of transcription-3 (STAT3) is a transcription factor that is activated by estrogen, translocates to the nucleus, and induces the transcription of neuroprotective genes, such as bcl-2. We determined whether estradiol increases STAT3 activation in female rat brain after focal cerebral ischemia and whether STAT3 activation contributes to estradiol-mediated neuroprotection against ischemic brain injury. Ovariectomized (OVX) female rats with and without estradiol replacement were subjected to 2 h of middle cerebral artery occlusion (MCAO), and phosphorylated STAT3 (P-STAT3) and total STAT3 (T-STAT3) were quantified by Western blot analysis at 3 and 22 h of reperfusion. STAT3 activation was colocalized with neuronal and survival markers microtubule-associated protein 2 (MAP2) and Bcl-2 using immunohistochemistry. Infarct size was measured at 22 h after MCAO in estradiol-treated OVX animals in the presence and absence of STAT3 inhibitor cucurbitacin I (JSI-124) using 2,3,5-triphenyltetrazolium chloride staining. Estradiol increased P-STAT3 in the ischemic cortex cytosolic fraction at 3 h after MCAO without affecting T-STAT3. This was associated with increased P-STAT3 in the nuclear fraction, which remained elevated at 22 h after MCAO. The nuclear P-STAT3 colocalized with MAP2 and Bcl-2 within the peri-infarct zone. The P-STAT3 inhibitor JSI-124 abolished the protective effect of estradiol without affecting infarct size in untreated OVX rats. We conclude that estradiol increases STAT3 phosphorylation in neurons after MCAO and that STAT3 activation plays an important role in estradiol-mediated neuroprotection.
Collapse
Affiliation(s)
- Suzan Dziennis
- Departments of Anesthesiology and Peri-Operative Medicine and
| | - Taiping Jia
- Physiology and Pharmacology, Oregon Health & Science University, Portland, Oregon 97239-3098
| | - Oline K. Rønnekleiv
- Departments of Anesthesiology and Peri-Operative Medicine and
- Physiology and Pharmacology, Oregon Health & Science University, Portland, Oregon 97239-3098
| | - Patricia D. Hurn
- Departments of Anesthesiology and Peri-Operative Medicine and
- Physiology and Pharmacology, Oregon Health & Science University, Portland, Oregon 97239-3098
| | - Nabil J. Alkayed
- Departments of Anesthesiology and Peri-Operative Medicine and
- Physiology and Pharmacology, Oregon Health & Science University, Portland, Oregon 97239-3098
| |
Collapse
|
365
|
Ardelt AA, Anjum N, Rajneesh KF, Kulesza P, Koehler RC. Estradiol augments peri-infarct cerebral vascular density in experimental stroke. Exp Neurol 2007; 206:95-100. [PMID: 17509567 PMCID: PMC2045124 DOI: 10.1016/j.expneurol.2007.04.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2006] [Revised: 02/21/2007] [Accepted: 04/07/2007] [Indexed: 11/20/2022]
Abstract
Peri-infarct increase of vascular density has been observed in animals and in humans with ischemic stroke. Increased peri-infarct vascular density correlates with improved functional outcome after stroke. We hypothesized that pre-treatment with estradiol will increase post-ischemic peri-infarct capillary density in a rat model of transient ischemic stroke. Estradiol, compared to placebo, augmented post-ischemic peri-infarct vascular density by 22% 10 days after stroke. Recovery of forelimb function was not improved with estradiol treatment on day three and nine post-stroke. Loss of estradiol may limit repair in the peri-infarct region by limiting angiogenesis, but functional significance in stroke recovery requires further investigation.
Collapse
Affiliation(s)
- Agnieszka A Ardelt
- University of Alabama at Birmingham, Department of Neurology, Comprehensive Stroke Center, 1813 6th Avenue South, Suite M226 RWUH, Birmingham, AL 35294, USA.
| | | | | | | | | |
Collapse
|
366
|
Chen SH, Huang KF, Lin MT, Chang FM. Human umbilical cord blood cells or estrogen may be beneficial in treating heatstroke. Taiwan J Obstet Gynecol 2007; 46:15-25. [PMID: 17389184 DOI: 10.1016/s1028-4559(08)60101-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
This current review summarized animal models of heatstroke experimentation that promote our current knowledge of therapeutic effects on cerebrovascular dysfunction, coagulopathy, and/or systemic inflammation with human umbilical cord blood cells (HUCBCs) or estrogen in the setting of heatstroke. Accumulating evidences have demonstrated that HUCBCs provide a promising new therapeutic method against neurodegenerative diseases, such as stroke, traumatic brain injury, and spinal cord injury as well as blood disease. More recently, we have also demonstrated that post- or pretreatment by HUCBCs may resuscitate heatstroke rats with by reducing circulatory shock, and cerebral nitric oxide overload and ischemic injury. Moreover, CD34+ cells sorted from HUCBCs may improve survival by attenuating inflammatory, coagulopathy, and multiorgan dysfunction during experimental heatstroke. Many researchers indicated pro- (e.g. tumor necrosis factor-alpha [TNF-alpha]) and anti-inflammatory (e.g. interleukin-10 [IL-10]) cytokines in the peripheral blood stream correlate with severity of circulatory shock, cerebral ischemia and hypoxia, and neuronal damage occurring in heatstroke. It has been shown that intravenous administration of CD34+ cells can secrete therapeutic molecules, such as neurotrophic factors, and attenuate systemic inflammatory reactions by decreasing serum TNF-alpha but increasing IL-10 during heatstroke. Another line of evidence has suggested that estrogen influences the severity of injury associated with cerebrovascular shock. Recently, we also successfully demonstrated estrogen resuscitated heatstroke rats by ameliorating systemic inflammation. Conclusively, HUCBCs or estrogen may be employed as a beneficial therapeutic strategy in prevention and repair of cerebrovascular dysfunction, coagulopathy, and/or systemic inflammation during heatstroke.
Collapse
Affiliation(s)
- Sheng-Hsien Chen
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Department of Obstetrics and Gynecology, Chi Mei Medical Center, Tainan, Taiwan
| | | | | | | |
Collapse
|
367
|
Huang Y, Huang YL, Lai B, Zheng P, Zhu YC, Yao T. Raloxifene acutely reduces glutamate-induced intracellular calcium increase in cultured rat cortical neurons via inhibition of high-voltage-activated calcium current. Neuroscience 2007; 147:334-41. [PMID: 17543470 DOI: 10.1016/j.neuroscience.2007.04.037] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2006] [Revised: 03/09/2007] [Accepted: 04/04/2007] [Indexed: 12/20/2022]
Abstract
There is increasing evidence indicating that estrogen replacement therapy produces neuroprotective actions but has undesirable side effects on the reproductive system. Raloxifene is a selective estrogen receptor modulator that exerts estrogen agonist action in the brain while acting as an estrogen antagonist in the reproductive system. In the present study, we investigated whether raloxifene affected the glutamate-induced calcium (Ca2+) overload in rat cultured cortical neurons. The bulk cytosolic intracellular Ca2+ level was measured by using confocal microscopy with fluorescent Ca2+ probe fluo3. Whole-cell recording technique was used to observe the effects of raloxifene on N-methyl-D-aspartate (NMDA)-evoked and voltage-activated Ca2+ currents in cultured cortical neurons. Pre-exposure of cortical neurons to raloxifene (0.5 microM-10 microM) for 3 min attenuated intracellular Ca2+ increase induced by application of glutamate (300 microM) for 1 min. The action of raloxifene was reversible after washout. ICI 182,780 and thapsigargin did not block the action of raloxifene. In whole-cell recording experiments, raloxifene (10 microM) significantly reduced the amplitude of the high-voltage-activated Ca2+ current but had no effect on NMDA-evoked Ca2+ current. The present study demonstrates that raloxifene acutely reduces glutamate-induced intracellular Ca2+ increase probably via inhibition of high-voltage-activated calcium channels.
Collapse
Affiliation(s)
- Y Huang
- National Laboratory of Medical Neurobiology, Shanghai Medical College, Fudan University, Shanghai, China
| | | | | | | | | | | |
Collapse
|
368
|
Dhote VV, Balaraman R. Gender specific effect of progesterone on myocardial ischemia/reperfusion injury in rats. Life Sci 2007; 81:188-97. [PMID: 17585947 DOI: 10.1016/j.lfs.2007.05.010] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2006] [Revised: 04/27/2007] [Accepted: 05/01/2007] [Indexed: 11/24/2022]
Abstract
The study was designed to investigate the effect of progesterone and its gender based variation on myocardial ischemia/reperfusion (I/R) injury in rats. Adult Sprague Dawley rats were divided into vehicle treated reperfusion injury group male (I/R-M), female (I/R-F), ovariectomised (I/R-OVR) and progesterone treatment (I/R-M+PG, I/R-F+PG, I/R-OVR+PG) groups, respectively. I/R injury was produced by occluding the left descending coronary artery (LCA) for 1 h and followed by re-opening for 1 h. Progesterone (2 mg kg(-1) i.p.) was administered 30 min after induction of ischemia. Hemodynamic parameters (+/-dp/dt, MAP), heart rate, ST-segment elevation and occurrence of ventricular tachycardia (VT) were measured during the I/R period. The myocardial infarct area, oxidative stress markers, activities of myeloperoxidase (MPO) and creatine kinase (CK) were determined after the experiment along with the assessment of the effect on apoptotic activity by using DNA fragmentation analysis. Histological observations were carried out on heart tissue. Treatment with progesterone significantly (P<0.05) reduced infarct area, lipid peroxidation (LPO) level and activity of MPO in females (I/R-F+PG) as compared to ischemic females (I/R-F). Progesterone significantly (P<0.001, P<0.05) inhibited serum CK activity and incidences of VT in female rats. Superoxide dismutase (SOD) activity, reduced glutathione (GSH) levels were significantly elevated (P<0.05) in I/R-F+PG group. Internucleosomal DNA fragmentation was less in I/R-F+PG group when compared to I/R-F group. The ischemic male and ovariectomised (I/R-M and I/R-OVR) counterparts did not show any significant change after progesterone treatment. In conclusion, the cardioprotective effect of progesterone on myocardial I/R injury induced damage is based on gender of the animal. The protective effect could be mediated by attenuation of inflammation and its possible interaction with endogenous estrogen.
Collapse
Affiliation(s)
- Vipin V Dhote
- Pharmacy Department, The M.S. University of Baroda, Faculty of Technology and Engineering, Kalabhavan, Vadodara, 390001, Gujarat, India
| | | |
Collapse
|
369
|
Durukan A, Tatlisumak T. Acute ischemic stroke: overview of major experimental rodent models, pathophysiology, and therapy of focal cerebral ischemia. Pharmacol Biochem Behav 2007; 87:179-97. [PMID: 17521716 DOI: 10.1016/j.pbb.2007.04.015] [Citation(s) in RCA: 526] [Impact Index Per Article: 29.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2006] [Revised: 04/16/2007] [Accepted: 04/19/2007] [Indexed: 12/22/2022]
Abstract
Ischemic stroke is a devastating disease with a complex pathophysiology. Animal modeling of ischemic stroke serves as an indispensable tool first to investigate mechanisms of ischemic cerebral injury, secondly to develop novel antiischemic regimens. Most of the stroke models are carried on rodents. Each model has its particular strengths and weaknesses. Mimicking all aspects of human stroke in one animal model is not possible since ischemic stroke is itself a very heterogeneous disorder. Experimental ischemic stroke models contribute to our understanding of the events occurring in ischemic and reperfused brain. Major approaches developed to treat acute ischemic stroke fall into two categories, thrombolysis and neuroprotection. Trials aimed to evaluate effectiveness of recombinant tissue-type plasminogen activator in longer time windows with finer selection of patients based on magnetic resonance imaging tools and trials of novel recanalization methods are ongoing. Despite the failure of most neuroprotective drugs during the last two decades, there are good chances to soon have effective neuroprotectives with the help of improved preclinical testing and clinical trial design. In this article, we focus on various rodent animal models, pathogenic mechanisms, and promising therapeutic approaches of ischemic stroke.
Collapse
Affiliation(s)
- Aysan Durukan
- Department of Neurology, Helsinki University Central Hospital, Biomedicum Helsinki, POB 700, Haartmaninkatu 8, 00290 Helsinki, Finland.
| | | |
Collapse
|
370
|
Brann DW, Dhandapani K, Wakade C, Mahesh VB, Khan MM. Neurotrophic and neuroprotective actions of estrogen: basic mechanisms and clinical implications. Steroids 2007; 72:381-405. [PMID: 17379265 PMCID: PMC2048656 DOI: 10.1016/j.steroids.2007.02.003] [Citation(s) in RCA: 481] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2006] [Revised: 02/07/2007] [Accepted: 02/09/2007] [Indexed: 12/16/2022]
Abstract
Estrogen is an important hormone signal that regulates multiple tissues and functions in the body. This review focuses on the neurotrophic and neuroprotective actions of estrogen in the brain, with particular emphasis on estrogen actions in the hippocampus, cerebral cortex and striatum. Sex differences in the risk, onset and severity of neurodegenerative disease such as Alzheimer's disease, Parkinson's disease and stroke are well known, and the potential role of estrogen as a neuroprotective factor is discussed in this context. The review assimilates a complex literature that spans research in humans, non-human primates and rodent animal models and attempts to contrast and compare the findings across species where possible. Current controversies regarding the Women's Health Initiative (WHI) study, its ramifications, concerns and the new studies needed to address these concerns are also addressed. Signaling mechanisms underlying estrogen-induced neuroprotection and synaptic plasticity are reviewed, including the important concepts of genomic versus nongenomic mechanisms, types of estrogen receptor involved and their subcellular targeting, and implicated downstream signaling pathways and mediators. Finally, a multicellular mode of estrogen action in the regulation of neuronal survival and neurotrophism is discussed, as are potential future directions for the field.
Collapse
Affiliation(s)
- Darrell W Brann
- Institute of Molecular Medicine and Genetics, School of Medicine, Medical College of Georgia, Augusta, GA 30912, United States.
| | | | | | | | | |
Collapse
|
371
|
Simpkins JW, Dykens JA. Mitochondrial mechanisms of estrogen neuroprotection. ACTA ACUST UNITED AC 2007; 57:421-30. [PMID: 17512984 DOI: 10.1016/j.brainresrev.2007.04.007] [Citation(s) in RCA: 96] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2007] [Revised: 04/20/2007] [Accepted: 04/21/2007] [Indexed: 11/19/2022]
Abstract
Oxidative stress, bioenergetic failure and mitochondrial dysfunction are all implicated in the etiology of neurodegenerative diseases such as Alzheimer's disease (AD). The mitochondrial involvement in neurodegenerative diseases reflects the regulatory role mitochondrial failure plays in both necrotic cell death and apoptosis. The potent feminizing hormone, 17 beta-estradiol (E2), is neuroprotective in a host of cell and animal models of stroke and neurodegenerative diseases. The discovery that 17alpha-estradiol, an isomer of E2, is equally as neuroprotective as E2 yet is >200-fold less active as a hormone, has permitted development of novel, more potent analogs where neuroprotection is independent of hormonal potency. Studies of structure-activity relationships and mitochondrial function have led to a mechanistic model in which these steroidal phenols intercalate into cell membranes where they block lipid peroxidation reactions, and are in turn recycled. Indeed, the parental estrogens and novel analogs stabilize mitochondria under Ca(2+) loading otherwise sufficient to collapse membrane potential. The neuroprotective and mitoprotective potencies for a series of estrogen analogs are significantly correlated, suggesting that these compounds prevent cell death in large measure by maintaining functionally intact mitochondria. This therapeutic strategy is germane not only to sudden mitochondrial failure in acute circumstances, such as during a stroke or myocardial infarction, but also to gradual mitochondrial dysfunction associated with chronic degenerative disorders such as AD.
Collapse
Affiliation(s)
- James W Simpkins
- Department of Pharmacology and Neuroscience, Institute for Aging and Alzheimer's Disease Research, University of North Texas Health Science Center, 3500 Camp Bowie Boul., Fort Worth, TX 76102, USA.
| | | |
Collapse
|
372
|
Chen CH, Xue R, Zhang J, Li X, Mori S, Bhardwaj A. Effect of osmotherapy with hypertonic saline on regional cerebral edema following experimental stroke: a study utilizing magnetic resonance imaging. Neurocrit Care 2007; 7:92-100. [PMID: 17657661 DOI: 10.1007/s12028-007-0033-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
INTRODUCTION Hypertonic saline (HS) solutions are increasingly being utilized as osmotherapeutic agents for the treatment of cerebral edema associated with brain injury from diverse etiologies. METHODS In a rat model of permanent focal ischemia, we (1) determined the effect of HS therapy on regional brain water content with T(1)- and T(2)-weighted magnetic resonance imaging (MRI) and (2) tested the hypothesis that HS therapy modulates the expression of aquaporin-4 (AQP4) in the ischemic brain. RESULTS Halothane-anesthetized male Wistar rats were subjected to permanent middle cerebral artery occlusion (MCAO) and at 6 hr post-MCAO were treated with either continuous intravenous infusion of 0.9% saline (NS) or 7.5% HS for 18 hr. While lesion size measured on T(2)-weighted imaging did not differ between NS (580 +/- 217 mm(3); mean +/- SD) and HS (460 +/- 86 mm(3)) treatments, there was a correlation between T(2) values and tissue water content as determined by wet-to-dry ratio in the caudoputamen (CP) complex of ischemic core (r = 0.612, P < 0.05). There were significant differences in T(1) values with treatment in the ischemic cortex (NS: 2.08 +/- 0.13; HS: 1.78 +/- 0.20) and CP complex (NS: 2.09 +/- 0.14; HS: 1.77 +/- 0.22), but there was no correlation between T(2) values and regional brain tissue water content in the peri-infarct regions and the non-ischemic hemisphere. There were significant differences in AQP4 protein expression in the ischemic hemisphere between NS and HS-treated rats. CONCLUSIONS These data demonstrate that (1) T(2)-weighted MRI imaging correlates with tissue water content in the ischemic core but not in the peri-infarct regions, and (2) attenuation of ischemia-evoked cerebral edema involves the modulation of AQP4 channels in the brain.
Collapse
Affiliation(s)
- Chih-Hung Chen
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MA, USA
| | | | | | | | | | | |
Collapse
|
373
|
Kellert BA, McPherson RJ, Juul SE. A comparison of high-dose recombinant erythropoietin treatment regimens in brain-injured neonatal rats. Pediatr Res 2007; 61:451-5. [PMID: 17515870 DOI: 10.1203/pdr.0b013e3180332cec] [Citation(s) in RCA: 119] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Recombinant human erythropoietin (rEpo) is neuroprotective in neonatal models of hypoxic-ischemic brain injury. However, the optimal rEpo dose, dosing interval, and number of doses for reducing brain injury are still undetermined. We compared the neuroprotective efficacy of several subcutaneous rEpo treatment regimens. Seven-day-old rats underwent unilateral carotid ligation plus 90 min 8% hypoxia. Treatment began immediately after injury. Treatment regimens examined included 1, 3, or 7 daily subcutaneous injections of either 0 (vehicle), 2,500, 5,000, or 30,000 U/kg rEpo. Gross brain injury, neuronal apoptosis (TUNEL), and gliosis (glial fibrillary acidic protein) were assessed at 48 h or 1 wk post injury. Immunoreactive cells and brain injury were quantified for statistical comparison to vehicle controls. rEpo treatment reduced brain injury, apoptosis, and gliosis, in a dose-dependent U-shaped manner at both 48 h and 1 wk. Neither one injection of 2,500, seven injections of 5,000, or three injections of 30,000 U/kg rEpo were protective. Three doses of 5,000 and one dose of 30,000 U/kg rEpo were most protective at both time intervals. rEpo provides dose-dependent neuroprotection. Of the regimens tested, three doses of 5,000 U/kg was optimal because it provided maximal benefit with limited total exposure.
Collapse
Affiliation(s)
- Brian A Kellert
- Department of Pediatrics, University of Washington, Seattle, Washington 98195, USA
| | | | | |
Collapse
|
374
|
De Butte-Smith M, Nguyen AP, Zukin RS, Etgen AM, Colbourne F. Failure of estradiol to ameliorate global ischemia-induced CA1 sector injury in middle-aged female gerbils. Brain Res 2007; 1153:214-20. [PMID: 17462607 DOI: 10.1016/j.brainres.2007.03.082] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2007] [Revised: 03/16/2007] [Accepted: 03/21/2007] [Indexed: 11/29/2022]
Abstract
Global forebrain ischemia arising from brief occlusion of the carotid arteries in gerbils produces selective hippocampal CA1 neuronal loss. Pre-treatment with 17beta-estradiol ameliorates, in part, ischemia-induced damage in young animals. Because stroke and cardiac arrest are more likely to occur among elderly individuals, neuroprotective studies in older animals have compelling clinical relevance. We investigated whether estradiol would attenuate ischemia-induced hippocampal neuronal injury in middle-aged (12-14 months) male, intact female, ovariectomized (OVX) female and OVX females treated for 14 days with estradiol. Core temperature telemetry probes were also implanted at the time that estradiol was initiated. Ischemia was induced by bilateral occlusion of the common carotid arteries (5 min), during which time skull temperature was maintained under normothermic conditions. Estradiol blocked the modest spontaneous hyperthermia that normally follows ischemia. However, all four groups exhibited substantial neuronal cell loss in the CA1, assessed at 7 after ischemia. These findings indicate that estradiol pre-treatment under conditions that produce neuroprotection in young animals does not protect against ischemia-induced CA1 cell loss in middle-aged female gerbils.
Collapse
|
375
|
Toung TJK, Chen CH, Lin C, Bhardwaj A. Osmotherapy with hypertonic saline attenuates water content in brain and extracerebral organs. Crit Care Med 2007; 35:526-31. [PMID: 17205030 DOI: 10.1097/01.ccm.0000253309.44567.a6] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE Because of their beneficial effects in patients with hemorrhagic shock and multiple-system trauma, hypertonic saline solutions are increasingly being used perioperatively for volume resuscitation. Although the anti-edema effects of hypertonic saline on brain are well documented in a variety of brain injury paradigms, its effects on the water content on other organs has not been studied rigorously. In this study, we tested the hypothesis that a) hypertonic saline when given as an intravenous bolus and continuous infusion attenuates water content of small bowel, lung, and brain in rats without neuro-injury; and b) attenuation of stroke-associated increases in lung water is dependent on achieving a target serum osmolality. DESIGN Prospective laboratory animal study. SETTING Research laboratory in a teaching hospital. SUBJECTS Adult male Wistar rats. INTERVENTIONS In the first series of experiments, under controlled conditions of normoxia, normocarbia, and normothermia, spontaneously breathing, halothane-anesthetized (1.0-1.5%) adult male Wistar rats (280-320 g) were treated in a blinded randomized fashion with 7.5% hypertonic saline or 0.9% normal saline in a 8-mL/kg intravenous infusion for 3 hrs followed by a continuous intravenous infusion (1 mL/kg/hr) of 5% hypertonic saline or normal saline, respectively (n=10 each), for 48 hrs. A second group of rats were treated with continuous infusion only for 48 hrs of either 7.5% hypertonic saline or normal saline (1 mL/kg/hr) (n=10 each) without an intravenous bolus. Naïve rats served as controls (n=10). Tissue water content of small bowel, lung, and brain was determined by comparing the wet-to-dry ratios at the end of the experiment. In a second series of experiments, rats (n=94) were subjected to 2 hrs of transient middle cerebral artery occlusion by the intraluminal occlusion technique. At 6 hrs following middle cerebral artery occlusion, rats were treated in a blinded randomized fashion with a continuous intravenous infusion of normal saline, 3% hypertonic saline, or 7.5% hypertonic saline for 24, 48, 72, and 96 hrs. Surgical shams served as controls (n=7). Hypertonic saline was instituted as chloride/acetate mixture (50:50) in all experiments. Serum osmolality was determined at the end of the experiment in all animals. MEASUREMENTS AND MAIN RESULTS In rats without neuro-injury that received intravenous bolus followed by a continuous infusion, lung water content was significantly reduced with hypertonic saline (73.9+/-1.1%; 359+/-10 mOsm/L) (mean+/-sd) compared with normal saline treatment (76.1+/-0.53%; 298+/-4 mOsm/L) as was water content of small bowel (hypertonic saline, 69.1+/-5.8%; normal saline, 74.7+/-0.71%) and brain (hypertonic saline, 78.1+/-0.87%; normal saline, 79.2+/-0.38%) at 48 hrs. Stroke-associated increases in lung water content were attenuated with 7.5% hypertonic saline at all time points. There was a strong correlation between serum osmolality and attenuation of stroke-associated increases in lung water content (r=-.647) CONCLUSIONS Bowel, lung, and brain water content is attenuated with hypertonic saline when serum osmolality is >350 mOsm/L without adverse effect on mortality in animals with and without neuro-injury. Attenuation of water content of extracerebral organs with hypertonic saline treatment may have therapeutic implications in perioperative fluid management in patients with and without brain injury.
Collapse
Affiliation(s)
- Thomas J K Toung
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | | | | |
Collapse
|
376
|
Ragonese P, D'Amelio M, Savettieri G. Implications for estrogens in Parkinson's disease: an epidemiological approach. Ann N Y Acad Sci 2007; 1089:373-82. [PMID: 17261781 DOI: 10.1196/annals.1386.004] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Evidence from experimental and epidemiological studies suggests a role of sex hormones in the pathogenic process leading to neurodegenerative diseases, (i.e., Alzheimer's and Parkinson's disease). The effects of sexual steroid hormones are complex and vary with the events of women's fertile life. Estrogens are supposed to influence dopamine synthesis, metabolism, and transport; however, there is no consensus regarding the direction, locus, and mechanism of the effect of estrogens on the dopaminergic system. A neuroprotective effect of estrogens has been demonstrated in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-animal models of Parkinson's disease (PD). Epidemiological studies indicate gender differences regarding the onset and the prognosis of PD. Most of the analytical studies explored the relationship between PD and exogenous estrogens. Only three studies investigated the role of endogenous estrogens in the risk of developing PD. These studies reported an increased risk of PD in conditions causing an early reduction in endogenous estrogens (early menopause, reduced fertile life length). Longer cumulative length of pregnancies has also been associated with an increased PD risk. A lack of consensus still exists on the effect of the type of menopause (surgical vs. natural) on PD risk. Finally, the effect of postmenopausal estrogen replacement therapy is still debated. Inconsistencies across studies are in part explained by the complexity of the mechanisms of action of sexual hormones and by the paucity of analytical studies.
Collapse
Affiliation(s)
- Paolo Ragonese
- Dipartimento Universitario di Neuroscienze Cliniche, Università di Palermo, Via Gaetano La Loggia 1-90129 Palermo, Italy
| | | | | |
Collapse
|
377
|
Sandstrom NJ, Rowan MH. Acute pretreatment with estradiol protects against CA1 cell loss and spatial learning impairments resulting from transient global ischemia. Horm Behav 2007; 51:335-45. [PMID: 17239878 PMCID: PMC1907528 DOI: 10.1016/j.yhbeh.2006.12.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2006] [Revised: 11/22/2006] [Accepted: 12/01/2006] [Indexed: 10/23/2022]
Abstract
Estradiol can act to protect against hippocampal damage resulting from transient global ischemia, but little is known about the functional consequences of such neuroprotection. The present study examines whether acute estradiol administered prior to the induction of transient global ischemia protects against hippocampal cell death and deficits in performance on a spatial learning task. Ovariectomized female rats were primed with estradiol benzoate or oil vehicle 48 and 24 h prior to experiencing one of three durations of 4-vessel occlusion (0, 5, or 10 min). Performance on the cued and hidden platform versions of the Morris water maze was assessed 1 week following ischemia. On the cued platform task, neither hormone treatment nor ischemia significantly influenced acquisition. When tested on the hidden platform task, however, oil-treated rats exhibited impairments in spatial learning after either 5 or 10 min of ischemia while estradiol-treated rats showed no impairments after 5 min of ischemia and only mild impairments after 10 min of ischemia. Immediately following behavioral testing, rats were perfused and survival of CA1 pyramidal cells was assessed. Ischemia was associated with the loss of CA1 pyramidal cells but rats that received estradiol prior to ischemia showed less severe damage. Furthermore, the extent of cell loss was correlated with degree of spatial bias expressed on a probe trial following hidden platform training. These findings indicate that acute exposure to estradiol prior to ischemia is both neuroprotective and functionally protective.
Collapse
Affiliation(s)
- Noah J Sandstrom
- Department of Psychology, Williams College, 18 Hoxsey Street, Williamstown, MA 01267, USA.
| | | |
Collapse
|
378
|
Macrae IM, Carswell HV. Oestrogen and stroke: the potential for harm as well as benefit. Biochem Soc Trans 2007; 34:1362-5. [PMID: 17073819 DOI: 10.1042/bst0341362] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Epidemiological studies point to a beneficial influence of the female reproductive hormones on stroke risk in that women have a lower incidence of stroke prior to the menopause compared with men, but this difference weakens with age and stroke risk in women rises after the menopause. However, recent Women's Health Initiative trials in post-menopausal women report an increased stroke risk on hormone replacement therapy. An influence of gender is also apparent on stroke outcome in animal models: female rats exposed to transient MCA (middle cerebral artery) occlusion sustain less brain damage than age-matched males, with loss of protection following ovariectomy. The major hormone thought to be responsible for beneficial influences on stroke incidence and outcome is oestrogen, and a large preclinical literature now exists where exogenously administered oestrogen has been studied in male and ovariectomized female rats using a range of stroke models and outcome measures. Most of these studies administer oestrogen prior to the stroke, use a model of transient ischaemia followed by reperfusion and report a significant oestrogen-induced neuroprotection. However, in some studies where the MCA is permanently occluded, oestrogen pre-treatment in ovariectomized female rats has been shown to significantly exacerbate ischaemic damage. Therefore preclinical results demonstrate harmful as well as beneficial influences of oestrogen on the ischaemic brain, highlighting the need for further study to elucidate the mechanisms responsible for both detrimental and beneficial influences. Ultimately, this could lead to the development of new classes of oestrogenic compounds with improved risk/benefit profiles, designed to selectively activate pathways inducing only the beneficial effects of oestrogen in vivo.
Collapse
Affiliation(s)
- I M Macrae
- Division of Clinical Neuroscience, Wellcome Surgical Institute, University of Glasgow, Garscube Estate, Glasgow G61 1QH, UK.
| | | |
Collapse
|
379
|
Najt P, Nicoletti M, Chen HH, Hatch JP, Caetano SC, Sassi RB, Axelson D, Brambilla P, Keshavan MS, Ryan ND, Birmaher B, Soares JC. Anatomical measurements of the orbitofrontal cortex in child and adolescent patients with bipolar disorder. Neurosci Lett 2007; 413:183-6. [PMID: 17276600 PMCID: PMC3581335 DOI: 10.1016/j.neulet.2006.10.016] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2006] [Revised: 09/15/2006] [Accepted: 10/04/2006] [Indexed: 11/30/2022]
Abstract
Imaging studies indicate smaller orbitofrontal cortex (OFC) volume in mood disorder patients compared with healthy subjects. We sought to determine whether child and adolescent patients with bipolar disorder have smaller OFC volumes than healthy controls. Fourteen children and adolescents meeting DSM-IV criteria for bipolar disorder (six males and eight females with a mean age+/-S.D.=15.5+/-3.2 years) and 20 healthy controls (11 males and nine females with mean age+/-S.D.=16.9+/-3.8 years) were studied. Orbitofrontal cortex volume was measured using magnetic resonance imaging. Male bipolar patients had smaller gray matter volumes in medial (p=0.044), right medial (0.037) and right (p=0.032) lateral OFC subdivisions compared to male controls. In contrast, female patients had larger gray matter volumes in left (p=0.03), lateral (p=0.012), left lateral (p=0.007), and trends for larger volumes in right lateral and left medial OFC subdivisions compared with female controls. Male patients exhibit smaller gray matter volumes, while female patients exhibit larger volumes in some OFC sub-regions. Gender differences in OFC abnormalities may be involved in illness pathophysiology among young bipolar patients.
Collapse
Affiliation(s)
- Pablo Najt
- MOOD-CNS Program, Division of Mood and Anxiety Disorders, Department of Psychiatry, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- South Texas Veterans Health Care System, Audie L. Murphy Division, San Antonio, TX, USA
| | - Mark Nicoletti
- MOOD-CNS Program, Division of Mood and Anxiety Disorders, Department of Psychiatry, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- South Texas Veterans Health Care System, Audie L. Murphy Division, San Antonio, TX, USA
| | - Hua Hsuan Chen
- MOOD-CNS Program, Division of Mood and Anxiety Disorders, Department of Psychiatry, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Department of Radiology, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - John P. Hatch
- MOOD-CNS Program, Division of Mood and Anxiety Disorders, Department of Psychiatry, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Department of Orthodontics, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Sheila C. Caetano
- MOOD-CNS Program, Division of Mood and Anxiety Disorders, Department of Psychiatry, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- South Texas Veterans Health Care System, Audie L. Murphy Division, San Antonio, TX, USA
- Department of Psychiatry, University of Sao Paulo, Sao Paulo, Brazil
| | - Roberto B. Sassi
- Department of Psychiatry, University of Sao Paulo, Sao Paulo, Brazil
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - David Axelson
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Paolo Brambilla
- Department of Pathology and Experimental & Clinical Medicine, Section of Psychiatry, University of Udine, Udine, Italy
| | - Macheri S. Keshavan
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Neal D. Ryan
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Boris Birmaher
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jair C. Soares
- MOOD-CNS Program, Division of Mood and Anxiety Disorders, Department of Psychiatry, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- South Texas Veterans Health Care System, Audie L. Murphy Division, San Antonio, TX, USA
- Department of Radiology, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Corresponding author at: MOOD-CNS Program, Division of Mood and Anxiety Disorders, Department of Psychiatry (MC 7792), The University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229-3900, USA. Tel.: +1 210 562 5487; fax: +1 210 562 5485. (J.C. Soares)
| |
Collapse
|
380
|
Nijboer CHA, Groenendaal F, Kavelaars A, Hagberg HH, van Bel F, Heijnen CJ. Gender-specific neuroprotection by 2-iminobiotin after hypoxia-ischemia in the neonatal rat via a nitric oxide independent pathway. J Cereb Blood Flow Metab 2007; 27:282-92. [PMID: 16736041 DOI: 10.1038/sj.jcbfm.9600342] [Citation(s) in RCA: 105] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
We have shown earlier that 2-iminobiotin (2-IB) reduces hypoxia-ischemia (HI)-induced brain damage in neonatal rats, and presumed that inhibition of nitric oxide synthases (NOS) was the underlying mechanism. We now investigated the effect of 2-IB treatment in P7 rat pups to determine the role of gender and the neuroprotective mechanism. Pups were subjected to HI (occlusion of right carotid artery and 120 mins FiO(2) 0.08) and received subcutaneous (s.c.) 10 mg/kg 2-IB at 0, 12 and 24 h after hypoxia. After 6 weeks, neuronal damage was assessed histologically. We determined cerebral nitrite and nitrate (NO(x)) and nitrotyrosine, heat-shock protein 70, cytosolic cytochrome c, cleaved caspase 3, nuclear translocation of apoptosis-inducing factor (AIF) and the effect of 2-IB on NOS activity in cultured cells. 2-Iminobiotin treatment reduced long-term brain damage in female but not male rats. Unexpectedly, 2-IB treatment did not reduce cerebral NO(x) or nitrotyrosine levels, and did not inhibit NOS activity in vitro. The gender-dependent neuroprotective effect of 2-IB was reflected in inhibition of the HI-induced increase in cytosolic cytochrome c and cleaved caspase 3 in females only. Hypoxia-ischemia-induced activation of AIF was observed in males only and was not affected by 2-IB. Post-HI treatment with 2-IB provides gender-specific long- and short-term neuroprotection in female P7 rats via inhibition of the cytochrome c-caspase 3 neuronal death pathway. 2-Iminobiotin did not alter cerebral NO(x) nor inhibited NOS in intact cells. Therefore, we conclude that it is highly unlikely that the neuroprotective effect of 2-IB involves NOS inhibition.
Collapse
Affiliation(s)
- Cora H A Nijboer
- Laboratory for Psychoneuroimmunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | | | | | | | | |
Collapse
|
381
|
Abstract
Hormonal ligands for the nuclear receptor superfamily have at least two interacting mechanisms of action: 1) classical transcriptional regulation of target genes (genomic mechanisms); and 2) nongenomic actions that are initiated at the cell membrane, which could impact transcription. Although transcriptional mechanisms are increasingly well understood, membrane-initiated actions of these ligands are incompletely understood. Historically, this has led to a considerable divergence of thought in the molecular endocrine field. We have attempted to uncover principles of hormone action that are relevant to membrane-initiated actions of estrogens. There is evidence that the membrane-limited actions of hormones, particularly estrogens, involve the rapid activation of kinases and the release of calcium. Membrane actions of estrogens, which activate these rapid signaling cascades, can also potentiate nuclear transcription. These signaling cascades may occur in parallel or in series but subsequently converge at the level of modification of transcriptionally relevant molecules such as nuclear receptors and/or coactivators. In addition, other hormones or neurotransmitters may also activate cascades to crosstalk with estrogen receptor-mediated transcription. The idea of synergistic coupling between membrane-initiated and genomic actions of hormones fundamentally revises the paradigms of cell signaling in neuroendocrinology.
Collapse
Affiliation(s)
- Nandini Vasudevan
- Department of Biology, Pennsylvania State University, University Park, Pennsylvania 16802, USA.
| | | |
Collapse
|
382
|
Mehta SL, Manhas N, Raghubir R. Molecular targets in cerebral ischemia for developing novel therapeutics. ACTA ACUST UNITED AC 2007; 54:34-66. [PMID: 17222914 DOI: 10.1016/j.brainresrev.2006.11.003] [Citation(s) in RCA: 540] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2006] [Revised: 11/09/2006] [Accepted: 11/10/2006] [Indexed: 11/20/2022]
Abstract
Cerebral ischemia (stroke) triggers a complex series of biochemical and molecular mechanisms that impairs the neurologic functions through breakdown of cellular integrity mediated by excitotoxic glutamatergic signalling, ionic imbalance, free-radical reactions, etc. These intricate processes lead to activation of signalling mechanisms involving calcium/calmodulin-dependent kinases (CaMKs) and mitogen-activated protein kinases (MAPKs) such as extracellular signal-regulated kinase (ERK), p38, and c-Jun N-terminal kinase (JNK). The distribution of these transducers bring them in contact with appropriate molecular targets leading to altered gene expression, e.g. ERK and JNK mediated early gene induction, responsible for activation of cell survival/damaging mechanisms. Moreover, inflammatory reactions initiated at the neurovascular interface and alterations in the dynamic communication between the endothelial cells, astrocytes and neurons are thought to substantially contribute to the pathogenesis of the disease. The damaging mechanisms may proceed through rapid nonspecific cell lysis (necrosis) or by active form of cell demise (apoptosis or necroptosis), depending upon the severity and duration of the ischemic insult. A systematic understanding of these molecular mechanisms with prospect of modulating the chain of events leading to cellular survival/damage may help to generate the potential strategies for neuroprotection. This review briefly covers the current status on the molecular mechanisms of stroke pathophysiology with an endeavour to identify potential molecular targets such as targeting postsynaptic density-95 (PSD-95)/N-methyl-d-aspartate (NMDA) receptor interaction, certain key proteins involved in oxidative stress, CaMKs and MAPKs (ERK, p38 and JNK) signalling, inflammation (cytokines, adhesion molecules, etc.) and cell death pathways (caspases, Bcl-2 family proteins, poly (ADP-ribose) polymerase-1 (PARP-1), apoptosis-inducing factor (AIF), inhibitors of apoptosis proteins (IAPs), heat shock protein 70 (HSP70), receptor interacting protein (RIP), etc., besides targeting directly the genes itself. However, selecting promising targets from various signalling cascades, for drug discovery and development is very challenging, nevertheless such novel approaches may lead to the emergence of new avenues for therapeutic intervention in cerebral ischemia.
Collapse
Affiliation(s)
- Suresh L Mehta
- Division of Pharmacology, Central Drug Research Institute, Chatter Manzil Palace, POB-173, Lucknow-226001, India
| | | | | |
Collapse
|
383
|
Nijboer CHA, Kavelaars A, van Bel F, Heijnen CJ, Groenendaal F. Gender-Dependent Pathways of Hypoxia-Ischemia-Induced Cell Death and Neuroprotection in the Immature P3 Rat. Dev Neurosci 2007; 29:385-92. [PMID: 17762206 DOI: 10.1159/000105479] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2006] [Accepted: 12/22/2006] [Indexed: 11/19/2022] Open
Abstract
Previously, we demonstrated neuroprotection with 2-iminobiotin (2-IB) after cerebral hypoxia-ischemia (HI) in female, but not in male P7 rats. Given the different patterns of brain injury in more immature rats, we examined whether these gender differences could also be observed in P3 rats. HI was induced by unilateral carotid ligation and FiO2 reduction, followed by 2-IB administration. HSP70 protein expression and cytochrome c release from the mitochondria, markers of short-term outcome, were induced by HI to the same extent in male and female animals. However, reduction in HSP70 production and cytochrome c release by 2-IB was seen in female rats only. Long-term cerebral injury after HI, assessed with histology, was similar in male and female P3 rats, but long-term neuroprotection by 2-IB was observed in female rats only. In conclusion, 2-IB provides neuroprotection after cerebral HI in female, but not in male immature P3 rats.
Collapse
Affiliation(s)
- Cora H A Nijboer
- Department of Neonatology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | | | | | | |
Collapse
|
384
|
Chiappetta O, Gliozzi M, Siviglia E, Amantea D, Morrone LA, Berliocchi L, Bagetta G, Corasaniti MT. Evidence to Implicate Early Modulation of Interleukin‐1β Expression in the Neuroprotection Afforded by 17β‐Estradiol in Male Rats Undergone Transient Middle Cerebral Artery Occlusion. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2007; 82:357-72. [PMID: 17678971 DOI: 10.1016/s0074-7742(07)82019-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Neuroprotection exerted by 17beta-estradiol (17beta-E(2)) has been widely investigated in animal models of acute cerebral ischemia. Estrogens interact with intracellular receptors (ERalpha and ERbeta) to modulate the transcription of target genes, including those implicated in neuronal survival. Neuroprotection may also occur via interaction with ER-like membrane receptors mediating rapid, non-genomic, actions or via receptor-independent mechanisms. There is also evidence that blockade of inflammatory factors may represent an important mechanism involved in estrogenic neuroprotection. Here we investigate whether reduced brain damage by acute pharmacological treatment with 17beta-E(2) in male rats subjected to transient (2h) middle cerebral artery occlusion (tMCAo) involves modulation of interleukin-1beta (IL-1beta), a proinflammatory cytokine strongly implicated in the pathophysiology of ischemic stroke. Administration of 17beta-E(2) (0.2mg/kg, i.p., 1h before tMCAo) results in significant reduction of brain infarct volume, and this is reverted by the ER antagonist ICI 182,780 (0.25mg/kg, i.p.) administered 1h before 17beta-E(2). Two hours MCAo followed by 2-h reperfusion results in a significant, threefold increase of IL-1beta levels in the cortical tissue ipsilateral to the ischemic damage. Interestingly, a pretreatment with a neuroprotective dose of 17beta-E(2) attenuates the cytokine elevation and this appears to occur through ER activation. In addition, neuroprotection by 17beta-E(2) is accompanied by reduced cytochrome c translocation both in the striatum and in the cortex as revealed by Western blotting 3h after reperfusion. In conclusion, we report the original observation that neuroprotection exerted by 17beta-E(2) in a rat model of transient focal brain ischemia is accompanied by reduced cytochrome c translocation to the cytosol and involves early modulation of IL-1beta production.
Collapse
Affiliation(s)
- Olga Chiappetta
- Department of Pharmacobiology, University of Calabria, Via P. Bucci 87036 Arcavacata di Rende (CS), Italy
| | | | | | | | | | | | | | | |
Collapse
|
385
|
Abstract
Fever is a common occurrence in patients following brain and spinal cord injury (SCI). In intensive care units, large numbers of patients demonstrate febrile periods during the first several days after injury. Over the last several years, experimental studies have reported the detrimental effects of fever in various models of central nervous system (CNS) injury. Small elevations in temperature during or following an insult have been shown to worsen histopathological and behavioral outcome. Thus, the control of fever after brain or SCI may improve outcome if more effective strategies for monitoring and treating hyperthermia were developed. Because of the clinical importance of fever as a potential secondary injury mechanism, mechanisms underlying the detrimental effects of mild hyperthermia after injury have been evaluated. To this end, studies have shown that mild hyperthermia (>37 degrees C) can aggravate multiple pathomechanisms, including excitotoxicity, free radical generation, inflammation, apoptosis, and genetic responses to injury. Recent data indicate that gender differences also play a role in the consequences of secondary hyperthermia in animal models of brain injury. The observation that dissociations between brain and body temperature often occur in head-injured patients has again emphasized the importance of controlling temperature fluctuations after injury. Thus, increased emphasis on the ability to monitor CNS temperature and prevent periods of fever has gained increased attention in the clinical literature. Cooling blankets, body vests, and endovascular catheters have been shown to prevent elevations in body temperature in some patient populations. This chapter will summarize evidence regarding hyperthermia and CNS injury.
Collapse
Affiliation(s)
- W Dalton Dietrich
- Department of Neurological Surgery, Miami Project to Cure Paralysis, University of Miami, Miller School of Medicine, Miami, FL 33136, USA.
| | | |
Collapse
|
386
|
Dong YL, Yue Y, Liu FH, Lang SY, Zhang XC, Dai SL, Ge QS, Zuo PP. Treatment with phytoestrogen alpha-zearalanol might protect neurons of hippocampus in ovariectomized rats. Endocrine 2006; 30:249-54. [PMID: 17526935 DOI: 10.1007/s12020-006-0001-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2006] [Revised: 10/09/2006] [Accepted: 10/09/2006] [Indexed: 11/24/2022]
Abstract
Although neuroprotective effects of estrogen on postmenopausal women have been recognized, an associated increased incidence of uterine and breast tumors has jeopardized the clinical use of estrogen. This study was designed to evaluate the neuroprotective effects of a novel phytoestrogen alpha-zearalanol (alpha-ZAL), on ovariectomized (OVX) rats. Adult Wistar rats were ovariectomized or sham-operated and treatment with equivalent doses of 17beta-estradiol or alpha-ZAL for 5 wk. Uteruses have been weighted and stained by hematoxylin and eosin for morphology analysis. The expression of synaptophysin and parvalbumin in hippocampus were evaluated by immunohistochemistry assays. Our experiments indicated that the synaptophysin and parvalbumin-positive areas were significantly decreased in the OVX group compared to the sham group, alpha-ZAL or 17beta-estradiol administration can reverse the effects. Although alpha-ZAL and 17beta-estradiol treatments reconciled uterus weight loss which was induced by ovariectomy, the effect of alpha-ZAL was less than 17beta-estradiol. This result suggests that alpha-ZAL may effectively abate neurons loss in the hippocampus while slightly promoting weight gain of the uterus.
Collapse
Affiliation(s)
- Yi-Long Dong
- Institute of Basic Science, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China 100005
| | | | | | | | | | | | | | | |
Collapse
|
387
|
Krause DN, Duckles SP, Pelligrino DA. Influence of sex steroid hormones on cerebrovascular function. J Appl Physiol (1985) 2006; 101:1252-61. [PMID: 16794020 DOI: 10.1152/japplphysiol.01095.2005] [Citation(s) in RCA: 308] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The cerebral vasculature is a target tissue for sex steroid hormones. Estrogens, androgens, and progestins all influence the function and pathophysiology of the cerebral circulation. Estrogen decreases cerebral vascular tone and increases cerebral blood flow by enhancing endothelial-derived nitric oxide and prostacyclin pathways. Testosterone has opposite effects, increasing cerebral artery tone. Cerebrovascular inflammation is suppressed by estrogen but increased by testosterone and progesterone. Evidence suggests that sex steroids also modulate blood-brain barrier permeability. Estrogen has important protective effects on cerebral endothelial cells by increasing mitochondrial efficiency, decreasing free radical production, promoting cell survival, and stimulating angiogenesis. Although much has been learned regarding hormonal effects on brain blood vessels, most studies involve young, healthy animals. It is becoming apparent that hormonal effects may be modified by aging or disease states such as diabetes. Furthermore, effects of testosterone are complicated because this steroid is also converted to estrogen, systemically and possibly within the vessels themselves. Elucidating the impact of sex steroids on the cerebral vasculature is important for understanding male-female differences in stroke and conditions such as menstrual migraine and preeclampsia-related cerebral edema in pregnancy. Cerebrovascular effects of sex steroids also need to be considered in untangling current controversies regarding consequences of hormone replacement therapies and steroid abuse.
Collapse
Affiliation(s)
- Diana N Krause
- Department of Pharmacology, School of Medicine, University of California, Irvine, 92697-4625, USA.
| | | | | |
Collapse
|
388
|
Turgeon JL, Carr MC, Maki PM, Mendelsohn ME, Wise PM. Complex actions of sex steroids in adipose tissue, the cardiovascular system, and brain: Insights from basic science and clinical studies. Endocr Rev 2006; 27:575-605. [PMID: 16763155 DOI: 10.1210/er.2005-0020] [Citation(s) in RCA: 176] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Recent publications describing the results of the Women's Health Initiative (WHI) and other studies reporting the impact of hormone therapy on aging women have spurred reexamination of the broad use of estrogens and progestins during the postmenopausal years. Here, we review the complex pharmacology of these hormones, the diverse and sometimes opposite effects that result from the use of different estrogenic and progestinic compounds, given via different delivery routes in different concentrations and treatment sequence, and to women of different ages and health status. We examine our new and growing appreciation of the role of estrogens in the immune system and the inflammatory response, and we pose the concept that estrogen's interface with this system may be at the core of some of the effects on multiple physiological systems, such as the adipose/metabolic system, the cardiovascular system, and the central nervous system. We compare and contrast clinical and basic science studies as we focus on the actions of estrogens in these systems because the untoward effects of hormone therapy reported in the WHI were not expected. The broad interpretation and publicity of the results of the WHI have resulted in a general condemnation of all hormone replacement in postmenopausal women. In fact, careful review of the extensive literature suggests that data resulting from the WHI and other recent studies should be interpreted within the narrow context of the study design. We argue that these results should encourage us to perform new studies that take advantage of a dialogue between basic scientists and clinician scientists to ensure appropriate design, incorporation of current knowledge, and proper interpretation of results. Only then will we have a better understanding of what hormonal compounds should be used in which populations of women and at what stages of menopausal/postmenopausal life.
Collapse
Affiliation(s)
- Judith L Turgeon
- Department of Internal Medicine, Division of Endocrinology, Clinical Nutrition, and Vascular Medicine, University of California Davis, Davis, California 95616, USA.
| | | | | | | | | |
Collapse
|
389
|
Xu Y, Zhang W, Klaus J, Young J, Koerner I, Sheldahl LC, Hurn PD, Martínez-Murillo F, Alkayed NJ. Role of cocaine- and amphetamine-regulated transcript in estradiol-mediated neuroprotection. Proc Natl Acad Sci U S A 2006; 103:14489-94. [PMID: 16971488 PMCID: PMC1636703 DOI: 10.1073/pnas.0602932103] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2006] [Indexed: 01/17/2023] Open
Abstract
Estrogen reduces brain injury after experimental cerebral ischemia in part through a genomic mechanism of action. Using DNA microarrays, we analyzed the genomic response of the brain to estradiol, and we identified a transcript, cocaine- and amphetamine-regulated transcript (CART), that is highly induced in the cerebral cortex by estradiol under ischemic conditions. Using in vitro and in vivo models of neural injury, we confirmed and characterized CART mRNA and protein up-regulation by estradiol in surviving neurons, and we demonstrated that i.v. administration of a rat CART peptide is protective against ischemic brain injury in vivo. We further demonstrated binding of cAMP response element (CRE)-binding protein to a CART promoter CRE site in ischemic brain and rapid activation by CART of ERK in primary cultured cortical neurons. The findings suggest that CART is an important player in estrogen-mediated neuroprotection and a potential therapeutic agent for stroke and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Yun Xu
- Departments of *Anesthesiology and Critical Care Medicine, and
- Department of Neurology, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Wenri Zhang
- Departments of Anesthesiology and Perioperative Medicine, and
| | - Judith Klaus
- Departments of *Anesthesiology and Critical Care Medicine, and
| | - Jennifer Young
- Departments of Anesthesiology and Perioperative Medicine, and
| | - Ines Koerner
- Departments of Anesthesiology and Perioperative Medicine, and
| | - Laird C. Sheldahl
- Physiology and Pharmacology, Oregon Health and Science University, Portland, OR 97239; and
| | - Patricia D. Hurn
- Departments of *Anesthesiology and Critical Care Medicine, and
- Departments of Anesthesiology and Perioperative Medicine, and
- Physiology and Pharmacology, Oregon Health and Science University, Portland, OR 97239; and
| | | | - Nabil J. Alkayed
- Departments of *Anesthesiology and Critical Care Medicine, and
- Departments of Anesthesiology and Perioperative Medicine, and
- Physiology and Pharmacology, Oregon Health and Science University, Portland, OR 97239; and
| |
Collapse
|
390
|
Plamondon H, Morin A, Charron C. Chronic 17beta-estradiol pretreatment and ischemia-induced hippocampal degeneration and memory impairments: a 6-month survival study. Horm Behav 2006; 50:361-9. [PMID: 16815389 DOI: 10.1016/j.yhbeh.2006.04.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2006] [Revised: 04/20/2006] [Accepted: 04/23/2006] [Indexed: 11/23/2022]
Abstract
Exogenous administration of estrogen has been shown to significantly reduce ischemia-induced neuronal degeneration. However, the long-term impact of such treatment on neuronal protection and functional recovery remain largely unknown. The present study assessed the effects of a 15-day pretreatment with 17beta-estradiol on memory deficits and neuronal damage up to 6 months following a 10-min global ischemia in rats. Four groups of ovariectomized female rats [sham-operated and ischemic rats receiving a 15-day pretreatment of either the vehicle or 17beta-estradiol (100 microg/kg)] were tested. The 8-arm radial maze and object recognition tests served to evaluate the impact of 17beta-estradiol treatment on ischemia-induced spatial and recognition memory impairments, respectively. Testing in the radial maze was initiated at two distinct time intervals following reperfusion (7 and 120 days) to evaluate changes in memory functions over time. Our findings revealed long-lasting neuroprotective effects of 17beta-estradiol treatment on hippocampal CA1 pyramidal cells in ovariectomized ischemic rats (43.5% greater neuronal survival than observed in vehicle-treated ischemic animals). Importantly, this neuronal protection translated into significant improvements of recognition and spatial memory functions in estradiol-treated ischemic rats.
Collapse
Affiliation(s)
- Hélène Plamondon
- School of Psychology, University of Ottawa, 11, Marie Curie, Room 204 Vanier Hall, Ottawa, ON, Canada.
| | | | | |
Collapse
|
391
|
Chaovipoch P, Jelks KAB, Gerhold LM, West EJ, Chongthammakun S, Floyd CL. 17beta-estradiol is protective in spinal cord injury in post- and pre-menopausal rats. J Neurotrauma 2006; 23:830-52. [PMID: 16774470 DOI: 10.1089/neu.2006.23.830] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The neuroprotective effects of 17 beta -estradiol have been shown in models of central nervous system injury, including ischemia, brain injury, and more recently, spinal cord injury (SCI). Recent epidemiological trends suggest that SCIs in elderly women are increasing; however, the effects of menopause on estrogen-mediated neuroprotection are poorly understood. The objective of this study was to evaluate the effects of 17beta-estradiol and reproductive aging on motor function, neuronal death, and white matter sparing after SCI of post- and pre-menopausal rats. Two-month-old or 1- year-old female rats were ovariectomized and implanted with a silastic capsule containing 180 microg/mL of 17beta-estradiol or vehicle. Complete crush SCI at T8-9 was performed 1 week later. Additional animals of each age group were left ovary-intact but were spinal cord injured. The Basso, Beattie, Bresnahan (BBB) locomotor test was performed. Spinal cords were collected on post-SCI days 1, 7, and 21, and processed for histological markers. Administration of 17beta-estradiol to ovariectomized rats improved recovery of hind-limb locomotion, increased white matter sparing, and decreased apoptosis in both the post- and pre-menopausal rats. Also, ovary-intact 1-year-old rats did worse than ovary-intact 2-month-old rats, suggesting that endogenous estrogen confers neuroprotection in young rats, which is lost in older animals. Taken together, these data suggest that estrogen is neuroprotective in SCI and that the loss of endogenous estrogen-mediated neuroprotective seen in older rats can be attenuated with exogenous administration of 17beta-estradiol.
Collapse
Affiliation(s)
- Pimonporn Chaovipoch
- Department of Neurological Surgery, University of California, Davis, California 95616, USA.
| | | | | | | | | | | |
Collapse
|
392
|
Reglödi D, Lubics A, Kiss P, Lengvári I, Gaszner B, Tóth G, Hegyi O, Tamás A. Effect of PACAP in 6-OHDA-induced injury of the substantia nigra in intact young and ovariectomized female rats. Neuropeptides 2006; 40:265-74. [PMID: 16904179 DOI: 10.1016/j.npep.2006.06.001] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2006] [Revised: 06/06/2006] [Accepted: 06/10/2006] [Indexed: 11/30/2022]
Abstract
Pituitary adenylate cyclase activating polypeptide (PACAP) has neuroprotective effects in various neuronal cultures and in models of brain pathologies in vivo. Among others, it protects dopaminergic neurons in vitro, against 6-OHDA- and rotenone-induced injury. Recently, we have shown that PACAP reduces dopaminergic cell loss and ameliorates behavioral outcome following unilateral 6-OHDA-induced injury of the substantia nigra in male rats. However, after castration, PACAP led only to a slight amelioration of the behavioral symptoms. The aim of the present study was to investigate the degree of neuroprotection exerted by PACAP in female rats, using the same model. It was found that PACAP had no effect on the dopaminergic cell loss in intact female rats, only caused amelioration of certain acute behavioral signs. In contrast, PACAP effectively increased dopaminergic cell survival and decreased behavioral deficits in ovariectomized females. These results indicate that the neuroprotective effect of PACAP in a rat model of Parkinson's disease is gender-specific.
Collapse
Affiliation(s)
- D Reglödi
- Department of Anatomy, Neurohumoral Research Group of the Hungarian Academy of Sciences, University of Pécs, Medical School, 7624 Pécs, Szigeti u 12, Hungary.
| | | | | | | | | | | | | | | |
Collapse
|
393
|
Al Qteishat A, Gaffney JJ, Krupinski J, Slevin M. Hyaluronan expression following middle cerebral artery occlusion in the rat. Neuroreport 2006; 17:1111-4. [PMID: 16837837 DOI: 10.1097/01.wnr.0000227986.69680.20] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Hyaluronic acid, a major component of the brain extracellular matrix, is a regulator of angiogenesis, cell differentiation and migration. We used the rat middle cerebral artery occlusion model to show hyaluronan accumulation in stroke-affected areas. Using reverse transcription-polymerase chain reaction and Western blotting we showed up-regulation of hyaluronidase-1 and 2 between 1 h and 21 days after stroke. Hyaluronidase-1 was up-regulated earlier than hyaluronidase-2. The hyaladherins, receptor for hyaluronan-mediated motility and CD44 were also increased after stroke. Using immunohistochemistry, we showed association of hyaluronidases 1/2 and hyaladherins with neurons in the infarcted and peri-infarcted regions and hyaluronidase-1 with microvessels. Hyaluronan synthesis and degradation in the stroke hemisphere might have an impact on neuronal survival, angiogenesis and general tissue remodelling after stroke.
Collapse
Affiliation(s)
- Ahmed Al Qteishat
- The School of Biology, Chemistry and Health Science, Manchester Metropolitan University, Manchester, UK
| | | | | | | |
Collapse
|
394
|
Chen CH, Toung TJK, Sapirstein A, Bhardwaj A. Effect of duration of osmotherapy on blood-brain barrier disruption and regional cerebral edema after experimental stroke. J Cereb Blood Flow Metab 2006; 26:951-8. [PMID: 16306935 DOI: 10.1038/sj.jcbfm.9600248] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Osmotherapy is the cornerstone of medical management for cerebral edema associated with large ischemic strokes. We determined the effect of duration of graded increases in serum osmolality with mannitol and hypertonic saline (HS) on blood-brain barrier (BBB) disruption and regional cerebral edema in a well-characterized rat model of large ischemic stroke. Halothane-anesthetized adult male Wistar rats were subjected to transient (2-h) middle cerebral artery occlusion (MCAO) by the intraluminal occlusion technique. Beginning at 6 h after MCAO, rats were treated with either no intravenous fluids or a continuous intravenous infusion (0.3 mL/h) of 0.9% saline, 20% mannitol, 3% HS, or 7.5% HS for 24, 48, 72, and 96 h. In the first series of experiments, BBB permeability was quantified by the Evans blue (EB) extravasation method. In the second series of experiments, water content was assessed by comparing wet-to-dry weight ratios in six predetermined brain regions. Blood-brain barrier disruption was maximal in rats treated with 0.9% saline for 48 h, but did not correlate with increases in serum osmolality or treatment duration with osmotic agents. Treatment with 7.5% HS attenuated water content in the periinfarct regions and all subregions of the contralateral nonischemic hemisphere to a greater extent than mannitol did with no adverse effect on survival rates. These data show that (1) BBB integrity is not affected by the duration and degree of serum osmolality with osmotic agents, and (2) attenuation of increases in brain water content with HS to target levels >350 mOsm/L may have therapeutic implications in the treatment of cerebral edema associated with ischemic stroke.
Collapse
Affiliation(s)
- Chih-Hung Chen
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | | | | |
Collapse
|
395
|
Wang X, Dykens JA, Perez E, Liu R, Yang S, Covey DF, Simpkins JW. Neuroprotective effects of 17beta-estradiol and nonfeminizing estrogens against H2O2 toxicity in human neuroblastoma SK-N-SH cells. Mol Pharmacol 2006; 70:395-404. [PMID: 16614138 DOI: 10.1124/mol.106.022384] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Neuroprotective effects of estrogens have been shown in various in vitro and in vivo models, but the mechanisms underlying protection by estrogen are not clear. Mounting evidence suggests antioxidant effects contribute to the neuroprotective effects of estrogens. In the present study, we assessed the protective effects of estrogens against H2O2-induced toxicity in human neuroblastoma cells and the potential mechanisms involved in this protection. We demonstrate that 17beta-estradiol (17beta-E2) increases cell survival against H2O2 toxicity in human neuroblastoma cells. 17beta-E2 effectively reduced lipid peroxidation induced by 5-min H2O2 exposure. Furthermore, 17beta-E2 exerts the protective effects by maintaining intracellular Ca2+ homeostasis, attenuating ATP depletion, ablating mitochondrial calcium overloading, and preserving mitochondrial membrane potential. Two nonfeminizing estrogens, 17alpha- and ent-estradiol, were as effective as 17beta-E2 in increasing cell survival, alleviating lipid peroxidation, preserving mitochondrial function, and maintaining intracellular glutathione levels and Ca2+ homeostasis against H2O2 insult. Moreover, the estrogen receptor antagonist fulvestrant (ICI 182,780) did not block effects of 17beta-E2, but increased cell survival and blunted intracellular Ca2+ increases. However, these estrogens failed to reduce cytosolic reactive oxygen species, even at concentrations as high as 10 microM. In conclusion, estrogens exert protective effects against oxidative stress by inhibiting lipid peroxidation and subsequently preserving Ca2+ homeostasis, mitochondrial membrane potential, and ATP levels.
Collapse
Affiliation(s)
- Xiaofei Wang
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, 3500 Camp Bowie Blvd., Fort Worth, TX 76107, USA
| | | | | | | | | | | | | |
Collapse
|
396
|
Raval AP, Bramlett H, Perez-Pinzon MA. Estrogen preconditioning protects the hippocampal CA1 against ischemia. Neuroscience 2006; 141:1721-30. [PMID: 16777351 DOI: 10.1016/j.neuroscience.2006.05.016] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2005] [Revised: 04/30/2006] [Accepted: 05/09/2006] [Indexed: 01/06/2023]
Abstract
Estrogen is neuroprotective against ischemia in both in vivo and in vitro injury models. Because of the promising preclinical data on neuroprotection, the Women's Estrogen for Stroke Trial was initiated. The outcomes from this trial were, however, unsuccessful and questions emerged about the safety of chronic estrogen treatment in women. In contrast to the chronic estrogen treatment strategy, the present study aims to investigate: (1) the neuroprotective efficacy of single estrogen pretreatment/preconditioning; and (2) the existence of a similarity between estrogen- and ischemic preconditioning-induced neuroprotection against cerebral ischemia. The efficacy of estrogen was tested in an in vitro model of cerebral ischemia using hippocampal organotypic slice culture system. The hippocampal organotypic slice cultures were generated from female neonatal (9-11 days old) Sprague-Dawley rats. The slices were exposed to estradiol-17beta (0.5, 1, 5 nM) for various durations (1, 2 or 4 h) 48 h prior to ischemia (40 min of oxygen-glucose deprivation). For ischemic preconditioning, slices were exposed to sublethal oxygen-glucose deprivation (15 min), 48 h prior to lethal oxygen-glucose deprivation. Quantification of cell death in hippocampal CA1 region was conducted by using propidium iodide fluorescence staining technique. Results demonstrated that estrogen preconditioning significantly protects the hippocampal CA1 region against ischemia (P<0.001) and mimicked ischemic preconditioning-induced neuroprotection. The propidium iodide fluorescence values of estrogen preconditioning, ischemic preconditioning and ischemia groups were 21+/-2 (mean+/-S.E.M.) (1 nM; 2 h; n=15), 18+/-2 (5 nM; 4 h; n=12), 32+/-3 (n=8), 65+/-3 (n=27), respectively. Further, estrogen preconditioning initiated a calcium-mediated signaling pathway leading to protection of CA1 neurons against ischemia. Future investigations in estrogen preconditioning may suggest new estrogen regimens that avoid potential side effects of chronic estrogen treatment for stroke patients.
Collapse
Affiliation(s)
- A P Raval
- Cerebral Vascular Disease Research Center, Department of Neurology and Neuroscience Program (D4-5), P.O. Box 016960, University of Miami School of Medicine, Miami, FL 33101, USA.
| | | | | |
Collapse
|
397
|
Singh M, Dykens JA, Simpkins JW. Novel mechanisms for estrogen-induced neuroprotection. Exp Biol Med (Maywood) 2006; 231:514-21. [PMID: 16636299 DOI: 10.1177/153537020623100505] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Estrogens are gonadal steroid hormones that are present in the circulation of both males and females and that can no longer be considered within the strict confines of reproductive function. In fact, the bone, the cardiovascular system, and extrahypothalamic regions of the brain are now well-established targets of estrogens. Among the numerous aspects of brain function regulated by estrogens are their effects on mood, cognitive function, and neuronal viability. Here, we review the supporting evidence for estrogens as neuroprotective agents and summarize the various mechanisms that may be involved in this effect, focusing particularly on the mitochondria as an important target. On the basis of this evidence, we discuss the clinical applicability of estrogens in treating various age-related disorders, including Alzheimer disease and stroke, and identify the caveats that must be considered.
Collapse
Affiliation(s)
- Meharvan Singh
- Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, USA.
| | | | | |
Collapse
|
398
|
Liu HH, Payne HR, Wang B, Brady ST. Gender differences in response of hippocampus to chronic glucocorticoid stress: role of glutamate receptors. J Neurosci Res 2006; 83:775-86. [PMID: 16498614 DOI: 10.1002/jnr.20782] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Glucocorticoids (GC) play critical roles in the pathophysiological reactions to environmental stress. In brain, morphological changes were examined in hippocampal CA3 neurons with 2 weeks of chronic elevation of GC in male and female mice. Molecular correlates and underlying mechanisms paralleling these morphologic changes in hippocampus were investigated. Although the hippocampal neurons in the CA3 area in male mice atrophy with chronically elevated GC, female mice show minimal morphological changes with comparable GC regimens. These sexual morphological differences correlate with differences in the postsynaptic dense protein (PSD95) as well as the spectrum of glutamate receptors induced by GC treatment in male and female mice, including NMDA, AMPA, and KA receptors. These findings suggest that synaptic receptor composition is adapted to the unique physiological requirements of males and females and illuminate underlying mechanisms of GC/stress responses in the brain.
Collapse
Affiliation(s)
- Howard H Liu
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Illinois 60612, USA
| | | | | | | |
Collapse
|
399
|
Farooque M, Suo Z, Arnold PM, Wulser MJ, Chou CT, Vancura RW, Fowler S, Festoff BW. Gender-related differences in recovery of locomotor function after spinal cord injury in mice. Spinal Cord 2006; 44:182-7. [PMID: 16130019 DOI: 10.1038/sj.sc.3101816] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
STUDY DESIGN In order to study the role of gender in recovery, we induced a thoracic compression spinal cord injury (SCI) separately in 2-month-old male and female C57Bl/6 mice. OBJECTIVES We intended to assess effects of gender on recovery of hindlimb motor function and to correlate these with histomorphologic profiles of injured spinal cord tissue. METHODS Locomotor function was evaluated by three means: a modified locomotor scoring system for rodents, beam walking and computerized activity meter. Histology was analyzed by comparison of hematoxylin and eosin-stained perfused specimens. RESULTS Locomotor scores were 2.2+/-0.9 on day 1 in male mice, while, in contrast, they were significantly higher, 7.3+/-1.7, in females (P<0.02). On day 14 Basso, Beattie and Bresnahan scores were 9.5+/-2.2 in male mice and 16.0+/-2.2 in females (P<0.03). Terminal histology showed that the spinal cord architecture was relatively better preserved in female mice and that the extent of necrosis and infiltration of inflammatory cells was less compared to males. SETTING Neurobiology Research Laboratory of University of Kansas Medical School in US Department of Veterans Affairs Medical Center, Kansas City, Missouri. CONCLUSION We found that the severity of the initial injury as well as the ultimate recovery of motor function after SCI is significantly influenced by gender, being remarkably better in females. The mechanism(s) of neuroprotection in females, although not yet elucidated, may be associated with the effects of estrogen on pathophysiological processes (blood flow, leukocyte migration inhibition, antioxidant properties, and inhibition of apoptosis). SPONSORSHIP Medical Research, US Department of Veterans Affairs, the Christopher Reeve Paralysis Foundation and NIH.
Collapse
Affiliation(s)
- M Farooque
- Neurobiology Research Laboratory, Department of Veterans Affairs Medical Center, Kansas City, MO 64128, USA
| | | | | | | | | | | | | | | |
Collapse
|
400
|
Kokiko ON, Murashov AK, Hoane MR. Administration of raloxifene reduces sensorimotor and working memory deficits following traumatic brain injury. Behav Brain Res 2006; 170:233-40. [PMID: 16580743 DOI: 10.1016/j.bbr.2006.02.026] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2005] [Revised: 01/24/2006] [Accepted: 02/20/2006] [Indexed: 11/29/2022]
Abstract
Hormonal differences between males and females have surfaced as a crucial component in the search for effective treatments after experimental models of traumatic brain injury (TBI). Recent findings have shown that selective estrogen receptor modulators (SERMs) may have therapeutic benefit. The present study examined the effects of raloxifene, a SERM, on functional recovery after bilateral cortical contusion injury (bCCI) or sham procedure. Male rats received injections of raloxifene (3.0mg/kg, i.p.) or vehicle (1.0 ml/kg, i.p.) 15 min, 24, 48, 72, and 96 h after bCCI or sham procedure. Rats were tested on both sensorimotor (bilateral tactile removal and locomotor placing tests) and cognitive tests (reference and working memory in the Morris water maze). Raloxifene-treated animals showed a significant reduction in the initial magnitude of the deficit and facilitated the rate of recovery for the bilateral tactile removal test, compared to vehicle-treated animals. The raloxifene-treated animals also showed a significant improvement in the acquisition of working memory compared to vehicle-treated animals. However, raloxifene did not significantly improve the acquisition of reference memory or locomotor placing ability. Raloxifene treatment also did not result in a significant reduction in the size of the lesion cavity. Thus, the task-dependent improvements seen following raloxifene treatment do not appear to be the result of cortical neuroprotection. However, these results suggest that raloxifene improves functional outcome following bCCI and may present an interesting avenue for future research.
Collapse
Affiliation(s)
- Olga N Kokiko
- Restorative Neuroscience Laboratory, Department of Psychology, Southern Illinois University, Carbondale, IL 62901, USA.
| | | | | |
Collapse
|