51
|
Durham JL, Jordan KA, Devos MJ, Williams EK, Sandstrom NJ. Estradiol protects against hippocampal damage and impairments in fear conditioning resulting from transient global ischemia in mice. Brain Res 2012; 1443:64-74. [PMID: 22305144 DOI: 10.1016/j.brainres.2012.01.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2011] [Revised: 12/12/2011] [Accepted: 01/07/2012] [Indexed: 01/02/2023]
Abstract
Estradiol protects against hippocampal damage and some learning impairments resulting from transient global ischemia in rats. Here, we seek to validate a mouse model of transient global ischemia and evaluate the effects of estradiol on ischemia-induced hippocampal damage and behavioral impairments. Female C57Bl6/J mice were ovariectomized and implanted with estradiol- or oil-secreting capsules. One week later, mice experienced 15-min of 2-vessel occlusion (2-VO) or sham surgical procedures. Five days later, mice were exposed to a fear conditioning protocol in which a specific context and novel tone were paired with mild footshock. Twenty-four hours following conditioning, contextual fear was assessed by measuring freezing behavior in the conditioned context (in the absence of the tone). This was followed by assessment of cue fear by measuring freezing behavior to the conditioned tone presented in a new context. When tested in the conditioned context, oil-treated mice that experienced 2-VO exhibited a significant reduction in freezing behavior whereas estradiol-treated mice that experienced 2-VO showed no disruption in freezing behavior. Freezing behavior when presented with the conditioned tone was unaffected by either surgery or hormone treatment. These findings suggest that global ischemia causes impairments in performance on the hippocampally-dependent contextual fear task but not conditioned cue-based fear. Furthermore, estradiol prevented the ischemia-induced impairment in contextual fear conditioning. Fluoro-Jade (FJ) staining revealed neuronal degeneration throughout the dorsal hippocampus of mice that experienced 2-VO. Estradiol treatment reduced the number of FJ+ cells in CA1 and CA2, but not in CA3 or in the dentate gyrus. Together, these findings suggest that 15 min of global ischemia causes extensive hippocampal neurodegeneration and disrupts contextual fear conditioning processes in mice and that estradiol protects against these adverse effects.
Collapse
Affiliation(s)
- Jennah L Durham
- Department of Psychology, Williams College, Williamstown, MA 01267, USA
| | | | | | | | | |
Collapse
|
52
|
Chen Y, Wu X, Yu S, Fauzee NJS, Wu J, Li L, Zhao J, Zhao Y. Neuroprotective Capabilities of Tanshinone IIA against Cerebral Ischemia/Reperfusion Injury via Anti-apoptotic Pathway in Rats. Biol Pharm Bull 2012; 35:164-70. [DOI: 10.1248/bpb.35.164] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Yanlin Chen
- Department of Pathology, Chongqing Medical University
| | - Xuemei Wu
- Department of Pathology, Chongqing Medical University
| | - Shanshan Yu
- Department of Pathology, Chongqing Medical University
| | | | - Jingxian Wu
- Department of Pathology, Chongqing Medical University
| | - Lan Li
- Department of Pathophysiology, Chongqing Medical University
| | - Jing Zhao
- Department of Pathophysiology, Chongqing Medical University
| | - Yong Zhao
- Department of Pathology, Chongqing Medical University
| |
Collapse
|
53
|
Kipp M, Berger K, Clarner T, Dang J, Beyer C. Sex steroids control neuroinflammatory processes in the brain: relevance for acute ischaemia and degenerative demyelination. J Neuroendocrinol 2012; 24:62-70. [PMID: 21592237 DOI: 10.1111/j.1365-2826.2011.02163.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Sex steroids have been demonstrated as powerful compounds to protect neurones and neural tissue from neurotoxic challenges and during neurodegeneration. A multitude of cellular actions have been attributed to female gonadal steroid hormones, including the regulation of pro-survival and anti-apoptotic factors, bioenergetic demands and radical elimination, growth factor allocation and counteracting against excitotoxicity. In recent years, immune-modulatory and anti-inflammatory characteristics of oestrogen and progesterone have also come under scrutiny. To date, each of these physiological responses has been considered to be partially and selectively integrated in the mediation of steroid-mediated cell protection and tested in suitable animal models and in vitro systems. To what extent these individual effects contribute to the overall neural protection remains sketchy. One idea is that a battery of cellular mechanisms operates at the same time. On the other hand, interactions and the control of the brain-intrinsic and peripheral immune system may play an additional and perhaps pioneering function in this scenario, notwithstanding the importance of secondary adjuvant mechanisms. In the present review, we highlight neuroprotective effects of oestrogen and progesterone in two different disease models of the brain, namely acute ischaemic and demyelination damage, which represent the most common acute and degenerative neurological disorders in humans. Besides other inflammatory parameters, we discuss the idea that chemokine expression and signalling appear to be early hallmarks in both diseases and are positively affected by sex steroids. In addition, the complex interplay with local brain-resident immune-competent cells appears to be controlled by the steroid environment.
Collapse
Affiliation(s)
- M Kipp
- Institute of Neuroanatomy, RWTH Aachen University, Aachen, Germany
| | | | | | | | | |
Collapse
|
54
|
Al Sweidi S, Sánchez MG, Bourque M, Morissette M, Dluzen D, Di Paolo T. Oestrogen receptors and signalling pathways: implications for neuroprotective effects of sex steroids in Parkinson's disease. J Neuroendocrinol 2012; 24:48-61. [PMID: 21790809 DOI: 10.1111/j.1365-2826.2011.02193.x] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Parkinson's disease (PD) is an age-related neurodegenerative disorder with a higher incidence in the male population. In the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of PD, 17β-oestradiol but not androgens were shown to protect dopamine (DA) neurones. We report that oestrogen receptors (ER)α and β distinctly contribute to neuroprotection against MPTP toxicity, as revealed by examining the membrane DA transporter (DAT), the vesicular monoamine transporter 2 (VMAT2) and tyrosine hyroxylase in ER wild-type (WT) and knockout (ERKO) C57Bl/6 male mice. Intact ERKOβ mice had lower levels of striatal DAT and VMAT2, whereas ERKOα mice were the most sensitive to MPTP toxicity compared to WT and ERKOβ mice and had the highest levels of plasma androgens. In both ERKO mice groups, treatment with 17β-oestradiol did not provide neuroprotection against MPTP, despite elevated plasma 17β-oestradiol levels. Next, the recently described membrane G protein-coupled oestrogen receptor (GPER1) was examined in female Macaca fascicularis monkeys and mice. GPER1 levels were increased in the caudate nucleus and the putamen of MPTP-monkeys and in the male mouse striatum lesioned with methamphetamine or MPTP. Moreover, neuroprotective mechanisms in response to oestrogens transmit via Akt/glycogen synthase kinase-3 (GSK3) signalling. The intact and lesioned striata of 17β-oestradiol treated monkeys, similar to that of mice, had increased levels of pAkt (Ser 473)/βIII-tubulin, pGSK3 (Ser 9)/βIII-tubulin and Akt/βIII-tubulin. Hence, ERα, ERβ and GPER1 activation by oestrogens is imperative in the modulation of ER signalling and serves as a basis for evaluating nigrostriatal neuroprotection.
Collapse
Affiliation(s)
- S Al Sweidi
- Molecular Endocrinology and Genomic Research Center, CHUQ (CHUL), Quebec City, Canada
| | | | | | | | | | | |
Collapse
|
55
|
Khaksari M, Soltani Z, Shahrokhi N, Moshtaghi G, Asadikaram G. The role of estrogen and progesterone, administered alone and in combination, in modulating cytokine concentration following traumatic brain injury. Can J Physiol Pharmacol 2011; 89:31-40. [PMID: 21186375 DOI: 10.1139/y10-103] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Cytokines play an important role in the pathophysiology of traumatic brain injury (TBI). This study was designed to determine the effects of administering progesterone (P) and estrogen (E), alone and in combination, on brain water content, blood-brain barrier (BBB) disturbance, and brain level of cytokines following diffuse TBI. Ovariectomized rats were divided into 9 groups, treated with vehicle, E1, E2, P1, P2, E1+P1, E1+P2, E2+P1, and E2+P2. Levels of BBB disruption (5 h), cytokines, and water content (24 h) were evaluated after TBI induced by the Marmarou method. Physiological (E1 and P1) and pharmacological (E2 and P2) doses of estrogen and progesterone were administered 30 min after TBI. Water content in the E1+P2-treated group was higher than in the E1-treated group. The inhibitory effect of E2 on water content was reduced by adding progesterone. The inhibitory effect of E1 and E2 on Evans blue content was reduced by treatment with E1+P1 and E2+P2, respectively. The brain level of IL-1β was reduced in E1 and E2, after TBI. In the E2+P2-treated group, this level was higher than in the E2-treated group. The brain level of TGF-β was also elevated by the administration of progesterone and estrogen alone, and reduced when the hormones were administered in combination. In conclusion, a combined administration of progesterone and estrogen inhibited the decreasing effects of administration of progesterone and estrogen alone on water content and BBB disruption that mediated to change the proinflammatory cytokines.
Collapse
Affiliation(s)
- Mohammad Khaksari
- Neuroscience Research Center, Kerman University of Medical Sciences, Iran.
| | | | | | | | | |
Collapse
|
56
|
Abstract
Newborn mammals are totally dependent on maternal milk and care for survival. The mother's brain undergoes different behavioural, physiological and emotional adaptations that make the mother more likely to satisfy the demands of the offspring. Recent reports from our group show that, compared to nulliparous rats, lactation diminishes cell damage induced by excitotoxicity in the dorsal hippocampus of the dam after systemic or i.c. administration of kainic acid (KA) and the resulting motor seizures. Elevated levels of prolactin (PRL), oxytocin, progesterone and glucocorticoids are characteristics of lactation, and the pronounced fluctuation of these hormones occurring in this phase may play a role protecting the hippocampus. Indeed, PRL administration to ovariectomised rats significantly diminishes the deleterious effects of KA in the dorsal hippocampus and reduces the progression of KA-induced seizures. Thus, lactation is a natural model for neuroprotection because it effectively prevents acute and chronic cell damage of the hippocampus induced by excitotoxicity.
Collapse
Affiliation(s)
- T Morales
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, México.
| |
Collapse
|
57
|
Etgen AM, Jover-Mengual T, Zukin RS. Neuroprotective actions of estradiol and novel estrogen analogs in ischemia: translational implications. Front Neuroendocrinol 2011; 32:336-52. [PMID: 21163293 PMCID: PMC3080451 DOI: 10.1016/j.yfrne.2010.12.005] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2010] [Revised: 12/06/2010] [Accepted: 12/09/2010] [Indexed: 11/15/2022]
Abstract
This review highlights our investigations into the neuroprotective efficacy of estradiol and other estrogenic agents in a clinically relevant animal model of transient global ischemia, which causes selective, delayed death of hippocampal CA1 neurons and associated cognitive deficits. We find that estradiol rescues a significant number of CA1 pyramidal neurons that would otherwise die in response to global ischemia, and this is true when hormone is provided as a long-term pretreatment at physiological doses or as an acute treatment at the time of reperfusion. In addition to enhancing neuronal survival, both forms of estradiol treatment induce measurable cognitive benefit in young animals. Moreover, estradiol and estrogen analogs that do not bind classical nuclear estrogen receptors retain their neuroprotective efficacy in middle-aged females deprived of ovarian hormones for a prolonged duration (8weeks). Thus, non-feminizing estrogens may represent a new therapeutic approach for treating the neuronal damage associated with global ischemia.
Collapse
Affiliation(s)
- Anne M Etgen
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA.
| | | | | |
Collapse
|
58
|
Barouk S, Hintz T, Li P, Duffy AM, MacLusky NJ, Scharfman HE. 17β-estradiol increases astrocytic vascular endothelial growth factor (VEGF) in adult female rat hippocampus. Endocrinology 2011; 152:1745-51. [PMID: 21343256 PMCID: PMC3075938 DOI: 10.1210/en.2010-1290] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2010] [Accepted: 01/25/2011] [Indexed: 01/17/2023]
Abstract
Vascular endothelial growth factor (VEGF) is critical to angiogenesis and vascular permeability. It is also important in the endocrine system, in which VEGF mediates the vascular effects of estrogens in target tissues such as the uterus, a response attributed to an estrogen response element on the VEGF gene. Here we asked whether 17β-estradiol increases VEGF levels in the brain. We focused on the hippocampus, in which 17β-estradiol and VEGF both have important actions, and used immunocytochemistry to evaluate VEGF protein. VEGF immunoreactivity was compared in adult female rats sampled during the estrous cycle when serum levels of 17β-estradiol peak (proestrous morning) as well as when they are low (metestrous morning). In addition, adult rats were ovariectomized and compared after treatment with 17β-estradiol or vehicle. The results demonstrated that VEGF immunoreactivity was increased when serum levels of 17β-estradiol were elevated. Confocal microscopy showed that VEGF immunofluorescence was predominantly nonneuronal, often associated with astrocytes. Glial VEGF labeling was primarily punctate rather than diffuse and labile because glial VEGF immunoreactivity was greatly reduced if tissue sections were left in an aqueous medium overnight. We conclude that VEGF protein in normal female hippocampus is primarily nonneuronal rather than neuronal and suggest that glial VEGF immunoreactivity has been underestimated by past studies with other methods because there is a labile extracellular pool. We suggest that estrogens may exert actions on female hippocampal structure and function by increasing hippocampal VEGF.
Collapse
Affiliation(s)
- Sharon Barouk
- The Nathan Kline Institute, Orangeburg, New York 10962, USA
| | | | | | | | | | | |
Collapse
|
59
|
Yao J, Chen S, Cadenas E, Brinton RD. Estrogen protection against mitochondrial toxin-induced cell death in hippocampal neurons: antagonism by progesterone. Brain Res 2011; 1379:2-10. [PMID: 21134358 PMCID: PMC3200366 DOI: 10.1016/j.brainres.2010.11.090] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2010] [Revised: 11/06/2010] [Accepted: 11/29/2010] [Indexed: 02/06/2023]
Abstract
Previously we demonstrated that mitochondrial dysfunction plays a critical role in the pathogenesis of Alzheimer's disease. Further, we have shown that the neuroprotective effects of 17β-estradiol (E2) are dependent upon mitochondrial function. In the current study, we sought to identify mitochondrial sites of E2 action that mediate neuroprotection by assessing the efficacy of E2 to protect neurons against inhibitors of mitochondrial respiration which target specific complexes within the respiratory chain. Subsequently, the impact of progesterone (P4) on E2-induced prevention against mitochondrial toxins was investigated. Mitochondrial inhibitors, rotenone, 3-NPA, antimycin, KCN, and oligomycin, exhibited concentration dependent toxicity in primary hippocampal neurons. The concentration inducing 30% cell death (LD30) was selected for analyses assessing the neuroprotective efficacy of ovarian hormones (E2 and P4). Pretreatment of hippocampal neurons with E2 significantly protected against 3-NPA (7.5mM) and antimycin (125 μM) induced cell death and was moderately neuroprotective against rotenone (3 μM). E2 was ineffective against KCN and oligomycin-induced cell death. Pretreatment with P4 was without effect against these mitochondrial inhibitors. Co-administration of P4 with E2 abolished E2 induced neuroprotection against 3-NPA and antimycin. Additional metabolic analyses indicated that E2 and P4 separately increased mitochondrial respiratory capacity whereas the co-administration of E2 and P4 resulted in diminished mitochondrial respiration. These findings indicate that E2 protects against mitochondrial toxins that target complexes I, II and III whereas P4 was without effect. The data also predict that continuous combined co-administration of estrogen and progesterone common to many hormone therapy regimens is unlikely to prevent the deficits in mitochondrial function.
Collapse
Affiliation(s)
- Jia Yao
- Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA 90033, USA
| | | | | | | |
Collapse
|
60
|
Kim JY, Jeong HY, Lee HK, Yoo JK, Bae K, Seong YH. Protective effect of Ilex latifolia, a major component of "kudingcha", against transient focal ischemia-induced neuronal damage in rats. JOURNAL OF ETHNOPHARMACOLOGY 2011; 133:558-564. [PMID: 21029769 DOI: 10.1016/j.jep.2010.10.037] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2010] [Revised: 10/15/2010] [Accepted: 10/18/2010] [Indexed: 05/30/2023]
Abstract
AIMS OF THE STUDY Ilex latifolia (Aquifoliaceae), a primary component of "kudingcha", has been used in Chinese folk medicine to treat various kinds of diseases including headaches, inflammatory diseases, and cardiac ischemic injury. The present study investigated the protective effect of the ethanol extract of Ilex latifolia against transient, focal, ischemia-induced neuronal damage. MATERIALS AND METHODS Transient focal ischemia was induced by 2 h middle cerebral artery occlusion followed by 24 h reperfusion (MCAO/reperfusion) in rats. After MCAO/reperfusion, brain infarction and neuronal death were measured by triphenyltetrazolium chloride and hematoxylin and eosin staining, respectively. Glutathione concentration and lipid peroxidation rate were measured. The expression levels of phosphorylated mitogen activated proteins kinases (MAPKs), cyclooxygenase 2 (COX-2), and anti-apoptotic and pro-apoptotic proteins were detected by Western blot. RESULTS Ilex latifolia (50-200 mg/kg) significantly reduced MCAO/reperfusion-induced infarction and edema formation, neurological deficits, and brain cell death. Depletion of glutathione level and lipid peroxidation induced by MCAO/reperfusion were inhibited by administration of Ilex latifolia. The increase of phosphorylated MAPKs, COX-2, and proapoptotic proteins and the decrease of antiapoptotic protein in MCAO/reperfusion rats were significantly inhibited by treatment with Ilex latifolia. CONCLUSION Ilex latifolia ameliorated ischemic injury induced by MCAO/reperfusion in rats, and this neuroprotective effect might be associated with its anti-apoptotic effect, resulting from anti-oxidative and anti-inflammatory actions.
Collapse
Affiliation(s)
- Joo Youn Kim
- College of Veterinary Medicine, Chungbuk National University, 410, Sungbong-ro, Heungduk-gu, Cheongju, Chungbuk 361-763, South Korea
| | | | | | | | | | | |
Collapse
|
61
|
Melcangi RC, Garcia-Segura LM. Sex differences in the injured brain. Horm Mol Biol Clin Investig 2011; 7:385-91. [DOI: 10.1515/hmbci.2011.112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2011] [Accepted: 08/15/2011] [Indexed: 11/15/2022]
Abstract
AbstractObservations obtained in human and in experimental models clearly demonstrate sex differences in degenerative events occurring in the central nervous system. The present review focuses on potential factors that may contribute to these sex-dimorphic features; in particular, morphological organization of the central nervous system and functional influence by neuroactive steroids, genes, and immune system are considered.
Collapse
|
62
|
Barda G, Ben-Haroush A, Barkat J, Malinger G, Luria O, Golan A, Bar J. Effect of vaginal progesterone, administered to prevent preterm birth, on impedance to blood flow in fetal and uterine circulation. ULTRASOUND IN OBSTETRICS & GYNECOLOGY : THE OFFICIAL JOURNAL OF THE INTERNATIONAL SOCIETY OF ULTRASOUND IN OBSTETRICS AND GYNECOLOGY 2010; 36:743-748. [PMID: 20196070 DOI: 10.1002/uog.7606] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 02/03/2010] [Indexed: 05/28/2023]
Abstract
OBJECTIVE To evaluate the effect on the maternal and fetal circulation of progesterone administered to prevent preterm birth. METHODS We used an observational cohort study design. The study group included 44 women at 18-32 weeks' gestation who presented with an episode of preterm labor, with or without history of delivery before 34 weeks' gestation, or an incidental finding of short cervix (≤ 25 mm). Doppler flow assessment of the umbilical artery, fetal middle cerebral artery and uterine arteries was performed before and 24 h after vaginal administration of progesterone. RESULTS Seventeen (38.6%) women gave birth before term, but only nine (20.4%) did so before 34 weeks' gestation. Following progesterone treatment, there was a statistically significant decrease in the pulsatility index of the fetal middle cerebral artery (mean reduction, 18.2%; mean change in pulsatility index, 0.44 (95% CI, 0.25-0.63), P < 0.001), with no changes in the other vessels. Comparison of the women who gave birth before with those who delivered at term yielded no significant differences in Doppler flow parameters in any vessel examined, either before or after progesterone treatment. CONCLUSION Treatment with vaginal progesterone is associated with a lower pulsatility index in the fetal middle cerebral artery, suggesting a vasodilatory effect on the fetal circulation.
Collapse
Affiliation(s)
- G Barda
- Department of Obstetrics and Gynecology, Edith Wolfson Medical Center, Holon, Israel
| | | | | | | | | | | | | |
Collapse
|
63
|
Baltmr A, Duggan J, Nizari S, Salt TE, Cordeiro MF. Neuroprotection in glaucoma - Is there a future role? Exp Eye Res 2010; 91:554-66. [PMID: 20800593 DOI: 10.1016/j.exer.2010.08.009] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2010] [Revised: 07/26/2010] [Accepted: 08/06/2010] [Indexed: 02/06/2023]
Abstract
In glaucoma, the major cause of global irreversible blindness, there is an urgent need for treatment modalities that directly target the RGCs. The discovery of an alternative therapeutic approach, independent of IOP reduction, is highly sought after, due to the indirect nature and limited effectiveness of IOP lowering therapy in preventing RGC loss. Several mechanisms have been implicated in initiating the apoptotic cascade in glaucomatous retinopathy and numerous drugs have been shown to be neuroprotective in animal models of glaucoma. These mechanisms and their potential treatment include excitotoxicity, protein misfolding, mitochondrial dysfunction, oxidative stress, inflammation and neurotrophin deprivation. All of these mechanisms ultimately lead to programmed cell death with loss of RGCs. In this article we summarize the mechanisms involved in glaucomatous disease, highlight the rationale for neuroprotection in glaucoma management and review current potential neuroprotective strategies targeting RGCs from the laboratory to the clinic.
Collapse
Affiliation(s)
- Abeir Baltmr
- Glaucoma and Retinal Neurodegeneration Research Group, Visual Neurosciences Department, University College London Institute of Ophthalmology, Bath Street, London EC1V 9EL, United Kingdom
| | | | | | | | | |
Collapse
|
64
|
Taylor LC, Puranam K, Gilmore W, Ting JPY, Matsushima G. 17beta-estradiol protects male mice from cuprizone-induced demyelination and oligodendrocyte loss. Neurobiol Dis 2010; 39:127-37. [PMID: 20347981 PMCID: PMC2891426 DOI: 10.1016/j.nbd.2010.03.016] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2009] [Revised: 02/25/2010] [Accepted: 03/19/2010] [Indexed: 12/15/2022] Open
Abstract
In addition to regulating reproductive functions in the brain and periphery, estrogen has tropic and neuroprotective functions in the central nervous system (CNS). Estrogen administration has been demonstrated to provide protection in several animal models of CNS disorders, including stroke, brain injury, epilepsy, Parkinson's disease, Alzheimer's disease, age-related cognitive decline and multiple sclerosis. Here, we use a model of toxin-induced oligodendrocyte death which results in demyelination, reactive gliosis, recruitment of oligodendrocyte precursor cells and subsequent remyelination to study the potential benefit of 17beta-estradiol (E2) administration in male mice. The results indicate that E2 partially ameliorates loss of oligodendrocytes and demyelination in the corpus callosum. This protection is accompanied by a delay in microglia accumulation as well as reduced mRNA expression of the pro-inflammatory cytokine, tumor necrosis factor alpha (TNFalpha), and insulin-like growth factor-1 (IGF-1). E2 did not significantly alter the accumulation of astrocytes or oligodendrocyte precursor cells, or remyelination. These data obtained from a toxin-induced, T cell-independent model using male mice provide an expanded view of the beneficial effects of estrogen on oligodendrocyte and myelin preservation.
Collapse
Affiliation(s)
- Lorelei C Taylor
- Curriculum in Neurobiology, University of North Carolina-CH, Chapel Hill, NC 27599
- UNC Neuroscience Center, University of North Carolina-CH, Chapel Hill, NC 27599
| | - Kasturi Puranam
- UNC Neuroscience Center, University of North Carolina-CH, Chapel Hill, NC 27599
| | - Wendy Gilmore
- Department of Neurology, University of Southern California, Los Angeles, CA 90033
| | - Jenny P-Y. Ting
- Curriculum in Neurobiology, University of North Carolina-CH, Chapel Hill, NC 27599
- Department of Microbiology and Immunology, University of North Carolina-CH, Chapel Hill, NC 27599
- UNC Neuroscience Center, University of North Carolina-CH, Chapel Hill, NC 27599
| | - G.K. Matsushima
- Curriculum in Neurobiology, University of North Carolina-CH, Chapel Hill, NC 27599
- Department of Microbiology and Immunology, University of North Carolina-CH, Chapel Hill, NC 27599
- UNC Neuroscience Center, University of North Carolina-CH, Chapel Hill, NC 27599
- Program for Molecular Biology and Biotechnology, University of North Carolina-CH, Chapel Hill, NC 27599
| |
Collapse
|
65
|
Tejadilla D, Cerbón M, Morales T. Prolactin reduces the damaging effects of excitotoxicity in the dorsal hippocampus of the female rat independently of ovarian hormones. Neuroscience 2010; 169:1178-85. [PMID: 20570717 DOI: 10.1016/j.neuroscience.2010.05.074] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2010] [Revised: 05/27/2010] [Accepted: 05/28/2010] [Indexed: 12/29/2022]
Abstract
We reported previously that lactation prevents the cell damage induced by kainic acid (KA) excitotoxicity in the CA1, CA3, and CA4 areas of the dorsal hippocampus compared to rats in diestrus phase, and hypothesize that pronounced fluctuations of hormones, such as ovarian steroids and prolactin (PRL), have a role in the neuroprotection of the dorsal hippocampus during lactation. PRL is thought to be involved in modulating neural excitability and seizure activity. To investigate actions of prolactin that minimize KA-induced cell damage in the hippocampus, female intact and ovariectomized (OVX) rats were treated for 4 days with a daily dose of 100 microg of prolactin or vehicle. On the third day of prolactin treatment, rats received a systemic dose of 7.5 mg/kg of KA and were sacrificed 48 h later. Immunostaining for Neu-N revealed a significant decrease in cell number in the CA1, CA3 and CA4 areas of intact or OVX, vehicle-treated rats after KA, whereas prolactin treatment prevented cell loss in the CA3 area of intact, and in the CA1, CA3, and CA4 of OVX rats. Fluoro-Jade C staining confirmed these observations. Kainate-induced seizure behavior progressed further in OVX rats, but was attenuated in prolactin-treated rats, both intact and OVX, compared to vehicle-treated rats. These data indicate that prolactin diminishes the damaging actions of excitotoxicity in the kainate model of epilepsy.
Collapse
Affiliation(s)
- D Tejadilla
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, Qro 76230, México
| | | | | |
Collapse
|
66
|
Lima FB, Bethea CL. Ovarian steroids decrease DNA fragmentation in the serotonin neurons of non-injured rhesus macaques. Mol Psychiatry 2010; 15:657-68. [PMID: 19823180 PMCID: PMC2874644 DOI: 10.1038/mp.2009.97] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2009] [Revised: 07/29/2009] [Accepted: 08/18/2009] [Indexed: 12/02/2022]
Abstract
We previously found that ovarian steroids promote neuroprotection in serotonin neurons by decreasing the expression of pro-apoptotic genes and proteins in the dorsal raphe nucleus of rhesus macaques, even in the absence of overt injury. In this study, we questioned whether these actions would lead to a reduction in DNA fragmentation in serotonin neurons. Ovariectomized (OVX) rhesus monkeys were implanted with silastic capsules that were empty (placebo) or containing estradiol (E), progesterone (P) or estradiol and progesterone (E+P) for 1 month. In all animals, eight levels of the dorsal raphe nucleus in a rostral-to-caudal direction were immunostained using the terminal deoxynucleotidyl transferase nick end labeling (TUNEL) method. Two staining patterns were observed, which are referred to as type I, with complete dark staining of the nucleus, and type II, with peripheral staining in the perinuclear area. A montage of the dorsal raphe was created at each level with a Marianas Stereology Microscope and Slidebook 4.2, and the TUNEL-positive cells were counted. In direct comparison with OVX animals, P treatment and E+P treatment significantly reduced the total number of TUNEL-positive cells (Mann-Whitney test, both treatments P=0.04) and E+P treatment reduced the number of TUNEL-positive cells per mm(3) (Mann-Whitney test, P=0.04). Double immunocytochemistry for TUNEL and tryptophan hydroxylase (TPH) indicated that DNA fragmentation was prominent in serotonin neurons. These data suggest that in the absence of ovarian steroids, a cascade of gene and protein expression leads to an increase in DNA fragmentation in serotonin neurons. Conversely, ovarian steroids have a neuroprotective role in the non-injured brain and prevent DNA fragmentation and cell death in serotonin neurons of nonhuman primates.
Collapse
Affiliation(s)
- Fernanda B. Lima
- Division of Reproductive Sciences Oregon National Primate Research Center Beaverton, Oregon, 97006, USA
| | - Cynthia L. Bethea
- Division of Reproductive Sciences Oregon National Primate Research Center Beaverton, Oregon, 97006, USA
- Division of Neuroscience Oregon National Primate Research Center Beaverton, Oregon, 97006, USA
| |
Collapse
|
67
|
Abstract
Clinical studies show that men are more likely to develop disorders affecting midbrain dopaminergic pathways, such as drug addiction and Parkinson's disease (PD). Although a great deal of focus has been given to the role of oestrogen in the maintenance of midbrain dopaminergic pathways, little is known about how testosterone influences these pathways. In the present study, we used stereological analysis of tyrosine hydroxylase-immunoreactive (TH-IR) cell bodies to determine how testosterone influences the dopaminergic cell bodies of the substantia nigra pars compacta (SNpc) and ventral tegmental area (VTA). Rats and mice were castrated at postnatal day (PN) 60, and these midbrain cell populations were counted on PN 90. One month after castration, TH-IR cell number had increased in the SNpc and VTA of rats and mice. Replacement with testosterone or the non-aromatisable analogue dihydrotestosterone (DHT) in castrated animals reduced TH-IR cell number in the SNpc and VTA in rats. In mice, the decrease of TH-IR cell number with testosterone or DHT replacement was observed only in the SNpc. The apparent increase in TH-IR neurone number after castration is not explained by an increase in TH expression because the number of nondopaminergic cells (TH-immunonegative, TH-IN) did not decrease proportionally after castration. TH-IN cell number did not change after castration or hormone replacement in rat or mouse SNpc or VTA. These findings suggest that testosterone may play a suppressive role in midbrain dopaminergic pathways.
Collapse
Affiliation(s)
- M L Johnson
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, USA
| | | | | | | | | | | |
Collapse
|
68
|
Johnson ML, Ho CC, Day AE, Walker QD, Francis R, Kuhn CM. Oestrogen receptors enhance dopamine neurone survival in rat midbrain. J Neuroendocrinol 2010; 22:226-37. [PMID: 20136693 PMCID: PMC3019761 DOI: 10.1111/j.1365-2826.2010.01964.x] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Previous findings in our laboratory and elsewhere have shown that ovariectomy of rats in adulthood attenuates cocaine-stimulated locomotor behaviour. Ovarian hormones enhance both cocaine-stimulated behaviour and increase dopamine overflow after psychomotor stimulants. The present study aimed to determine whether ovarian hormones have these effects in part by maintaining dopamine neurone number in the substantia nigra pars compacta (SNpc) and ventral tegmental area (VTA) and to investigate the roles of specific oestrogen receptors (ERs) in the maintenance of mesencephalic dopamine neurones. To accomplish this goal, we used unbiased stereological techniques to estimate the number of tyrosine hydroxylase-immunoreactive (TH-IR) cell bodies in midbrain regions of intact, ovariectomised and hormone-replaced female rats and mice. Animals received active or sham gonadectomy on postnatal day 60 and received vehicle, 17beta-oestradiol (E(2)) or selective ER agonists propyl-pyrazole-triol (PPT, ERalpha) or diarylpropionitrile (DPN, ERbeta) for 1 month post-surgery. In both rats and mice, ovariectomy reduced the number of TH-IR cells in the SNpc and VTA. Replacement with E(2), PPT or DPN prevented or attenuated the loss observed with ovariectomy in both rats and mice. An additional study using ER knockout mice revealed that adult female mice lacking ERalpha had fewer TH-IR cells in midbrain regions than wild-type mice, whereas mice lacking ERbeta had TH-IR cell counts comparable to wild-type. These findings suggest that, although both ER subtypes play a role in the maintenance of TH-IR cell number in the SNpc and VTA, ERalpha may play a more significant role.
Collapse
Affiliation(s)
- M L Johnson
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, USA
| | | | | | | | | | | |
Collapse
|
69
|
Abstract
OBJECTIVE The goal was to examine the association of gender with intraventricular hemorrhage (IVH) or periventricular leukomalacia (PVL) in a large national database. METHODS The National Inpatient Sample database was analyzed for the years 1998, 1999, 2001, 2002, and 2004 and its pediatrics version for the years 1997, 2000, and 2003. The National Inpatient Sample and its pediatrics version are annually collected databases for inpatient admissions from >1000 hospitals across the United States. We included all very low birth weight (VLBW) infants (birth weight [BW] of <1500 g) and excluded infants with major congenital anomalies. VLBW infants with IVH or PVL were identified by using International Classification of Diseases, Ninth Revision, diagnostic codes. We analyzed the data by using chi(2) and Fisher's exact tests to calculate odds ratios (ORs) and logistic regression analysis to control for possible confounders. RESULTS The data included 104 847 VLBW infants. With controlling for significant confounders, IVH was associated with male gender (15.9% vs 13.6%; adjusted OR [AOR]: 1.15 [95% confidence interval: 1.11-1.19]; P < .001). More boys than girls had severe IVH (38% vs 32.7%; AOR: 1.18 [95% confidence interval: 1.06-1.32]; P < .004). The increased rates of IVH and severe IVH in boys were significant in the BW subgroups of <1000 g and 1000 to 1499 g. The association of male gender with IVH and severe IVH was more significant in infants of 1000 to 1499 g (AOR: 1.19 vs 1.14; P = .006). The incidence of PVL in VLBW infants was 0.41%, and rates did not differ between genders (0.38% vs 0.43%; P = .42). CONCLUSIONS Compared with girls, VLBW male newborns are at greater risk to develop IVH and severe IVH but not PVL. The association of male gender with IVH or severe IVH is stronger with higher BWs.
Collapse
Affiliation(s)
- Mohamed A Mohamed
- George Washington University Medical Center, Newborn Services, 900 23rd St NW, Suite G-2092, Washington, DC 20037, USA.
| | | |
Collapse
|
70
|
Kramer M, Dang J, Baertling F, Denecke B, Clarner T, Kirsch C, Beyer C, Kipp M. TTC staining of damaged brain areas after MCA occlusion in the rat does not constrict quantitative gene and protein analyses. J Neurosci Methods 2010; 187:84-9. [PMID: 20064557 DOI: 10.1016/j.jneumeth.2009.12.020] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2009] [Revised: 12/27/2009] [Accepted: 12/29/2009] [Indexed: 10/20/2022]
Abstract
In models of ischemic stroke, TTC (2,3,5-triphenyltetrazolium chloride) staining is commonly applied for the fast and reliable visualization of hypoxic brain tissue and for defining the size of cerebral infarction and penumbra. Deciphering molecular processes of pathogenesis within the penumbra is of particular interest for the development of therapeutic strategies. The aim of this study was to assess whether TTC-stained tissues can easily and in a reliable quantitative manner be processed for further molecular and biochemical analyses. We applied phenol-based RNA isolation, protein lysis by conventional RIPA buffer, and combined RNA/protein isolation with NucleoSpinRNA/Protein-Kit. Gene and protein expression analyses were performed by RT-rtPCR and Western-blotting. Middle cerebral arteria occlusion (MCAO) in rats was performed following a standardized experimental procedure. After MCAO, TTC staining revealed massive cell death in cortical and sub-cortical areas. TTC processing did not affect the quality of tissue RNA and protein. The expression of housekeeping and regulatory genes and proteins revealed no difference between control and TTC-stained groups. The expression of known stroke-regulated genes such as TNFalpha and IL1beta revealed similar induction profiles after TTC staining as described in the literature. TTC staining allows the precise delineation of lesioned and primarily non-lesioned brain areas for subsequent dissection of selected tissue pieces for molecular analysis. Our study demonstrates that TTC-stained tissues in stroke animal models can be used for quantitative gene and protein expression analyses without constriction. Pathomechanisms of ongoing tissue damage within the penumbra region can now be investigated in detail.
Collapse
Affiliation(s)
- Martin Kramer
- Faculty of Medicine, Institute of Neuroanatomy, RWTH Aachen University, Wendlingweg 2, D-52074 Aachen, Germany
| | | | | | | | | | | | | | | |
Collapse
|
71
|
Yu SS, Zhao J, Zheng WP, Zhao Y. Neuroprotective effect of 4-hydroxybenzyl alcohol against transient focal cerebral ischemia via anti-apoptosis in rats. Brain Res 2010; 1308:167-75. [DOI: 10.1016/j.brainres.2009.10.037] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2009] [Revised: 10/12/2009] [Accepted: 10/14/2009] [Indexed: 02/04/2023]
|
72
|
Cuestas E, Bas J, Pautasso J. Sex differences in intraventricular hemorrhage rates among very low birth weight newborns. ACTA ACUST UNITED AC 2009; 6:376-82. [PMID: 19682665 DOI: 10.1016/j.genm.2009.06.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/11/2009] [Indexed: 11/16/2022]
Abstract
BACKGROUND The influence of male or female sex on newborn outcomes has been recognized for >30 years. Several studies have observed higher mortality and morbidity in males than in females. It is not clear how this sex difference is sustained in postnatal complications such as intraventricular hemorrhage (IVH), especially in very low birth weight (VLBW) newborns. OBJECTIVE This study examined possible sex-related differences in IVH rates among VLBW neonates. METHODS In a retrospective observational study conducted in Hospital Privado, Córdoba, Argentina, data from 332 consecutive VLBW newborns in a 12-year period were reviewed. Maternal factors, labor and delivery characteristics, and neonatal parameters, including the results of cranial ultrasound examination to detect IVH, were compared for males and females. Bivariate and multivariate logistic regression analyses were performed. RESULTS A total of 322 VLBW newborns were included, 168 males and 154 females. Compared with female neonates, male neonates had a higher risk of overall IVH (26.8% vs 9.7%; odds ratio [OR] = 3.4 [95% CI, 1.8-6.4]; P < 0.001) and for grades III or IV on the Papile scale (16.1% vs 1.9%; OR = 9.6 [95% CI, 2.9-32.5]; P < 0.001). In the multivariate logistic regression model, male sex sustained the association with a greater risk of IVH (OR = 6.8 [95% CI, 3.8-12.0]). CONCLUSIONS IVH was significantly associated with male sex in these VLBW newborns. Because other factors affect these differences, further research is required.
Collapse
Affiliation(s)
- Eduardo Cuestas
- Department of Pediatrics and Neonatology, Hospital Privado, School of Medicine, Catholic University of Córdoba, Córdoba, Argentina.
| | | | | |
Collapse
|
73
|
Traub ML, De Butte-Smith M, Zukin RS, Etgen AM. Oestradiol and insulin-like growth factor-1 reduce cell loss after global ischaemia in middle-aged female rats. J Neuroendocrinol 2009; 21:1038-44. [PMID: 19840235 PMCID: PMC2862345 DOI: 10.1111/j.1365-2826.2009.01927.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Whereas the ability of oestradiol and insulin-like growth factor (IGF)-1 to afford neuroprotection against ischaemia-induced neuronal death in young female and male rodents is well established, the impact of IGF-1 in middle-aged animals is largely unknown. The present study assessed the efficacy of oestradiol and IGF-1 with respect to reducing neuronal death after transient global ischaemia in middle-aged female rats after 8 weeks of hormone withdrawal. Rats were ovariohysterectomised and implanted 8 weeks later with an osmotic mini-pump delivering IGF-1 or saline into the lateral ventricle. Some rats also received physiological levels of oestradiol by subcutaneous pellet. Two weeks later, rats were subjected to global ischaemia or sham operation. Surviving hippocampal CA1 neurones were quantified. Ischaemia produced massive CA1 cell death compared to sham-operated animals, which was evident at 14 days. Significantly more neurones survived in animals treated with either oestradiol or IGF-1, but simultaneous treatment produced no additive effect. IGF-1, an endogenous growth factor, may be a clinically useful therapy in preventing human brain injury, with neuroprotective equivalence to oestradiol but without the harmful side-effects.
Collapse
Affiliation(s)
- Michael L. Traub
- Department of Obstetrics and Gynecology & Women’s Health, Albert Einstein College of Medicine of Yeshiva University, Jack and Pearl Resnick Campus, 1300 Morris Park Avenue, Bronx, NY USA 10461
| | - Maxine De Butte-Smith
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine of Yeshiva University, Jack and Pearl Resnick Campus, 1300 Morris Park Avenue, Bronx, NY USA 10461
| | - R. Suzanne Zukin
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine of Yeshiva University, Jack and Pearl Resnick Campus, 1300 Morris Park Avenue, Bronx, NY USA 10461
| | - Anne M. Etgen
- Department of Obstetrics and Gynecology & Women’s Health, Albert Einstein College of Medicine of Yeshiva University, Jack and Pearl Resnick Campus, 1300 Morris Park Avenue, Bronx, NY USA 10461
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine of Yeshiva University, Jack and Pearl Resnick Campus, 1300 Morris Park Avenue, Bronx, NY USA 10461
| |
Collapse
|
74
|
Bryan KJ, Mudd JC, Richardson SL, Chang J, Lee HG, Zhu X, Smith MA, Casadesus G. Down-regulation of serum gonadotropins is as effective as estrogen replacement at improving menopause-associated cognitive deficits. J Neurochem 2009; 112:870-81. [PMID: 19943850 DOI: 10.1111/j.1471-4159.2009.06502.x] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Declining levels of estrogen in women result in increases in gonadotropins such as luteinizing hormone (LH) through loss of feedback inhibition. LH, like estrogen, is modulated by hormone replacement therapy. However, the role of post-menopausal gonadotropin increases on cognition has not been evaluated. Here, we demonstrate that the down-regulation of ovariectomy-driven LH elevations using the gonadotropin releasing hormone super-analogue, leuprolide acetate, improves cognitive function in the Morris water maze and Y-maze tests in the absence of E2. Furthermore, our data suggest that these effects are independent of the modulation of estrogen receptors alpha and beta, or activation of CYP19 and StAR, associated with the production of endogenous E2. Importantly, pathways associated with improved cognition such as CaMKII and GluR1-Ser831 are up-regulated by leuprolide treatment but not by chronic long-term E2 replacement suggesting independent cognition-modulating properties. Our findings suggest that down-regulation of gonadotropins is as effective as E2 in modulating cognition but likely acts through different molecular mechanisms. These findings provide a potential novel protective strategy to treat menopause/age-related cognitive decline and/or prevent the development of AD.
Collapse
Affiliation(s)
- Kathryn J Bryan
- Department of Neurosciences, Case Western Reserve University, Cleveland, Ohio, USA
| | | | | | | | | | | | | | | |
Collapse
|
75
|
Heron PM, Turchan-Cholewo J, Bruce-Keller AJ, Wilson ME. Estrogen receptor alpha inhibits the estrogen-mediated suppression of HIV transcription in astrocytes: implications for estrogen neuroprotection in HIV dementia. AIDS Res Hum Retroviruses 2009; 25:1071-81. [PMID: 19886840 PMCID: PMC2828252 DOI: 10.1089/aid.2009.0065] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Many human immunodeficiency virus (HIV) proteins including Tat are produced by HIV-infected astrocytes and secreted into the brain resulting in extensive neuronal damage that contributes to the pathogenesis of HIV dementia. The neuroprotective hormone 17beta-estradiol (E2) is known to negatively regulate the HIV transcriptional promoter in human fetal astrocytes (SVGA cell line) in a Tat-dependent manner. In the present study we extended our investigation in HIV-infected SVGA cells and found a reduction in HIV p24 levels following E2 treatment in comparison to control. Although many E2-mediated events occur through estrogen receptor alpha (ERalpha), we found low levels of ERalpha mRNA and failed to detect ERalpha protein in SVGA cells. Paradoxically, when ERalpha was overexpressed the E2-mediated decrease in Tat transactivation of the promotor was prevented. To determine whether ERalpha expression is altered in the human brain following HIV infection, postmortum hippocampal tissue was obtained from cognitively normal HIV- and HIV+ patients, patients diagnosed with either mild cognitive/motor disorder (MCMD) or HIV-associated dementia (HAD). Immunohistochemistry and quantitative real-time PCR (qRT-PCR) for ERalpha and glial fibrillary acidic protein (GFAP) showed that ERalpha mRNA levels were not significantly different between groups, while GFAP increased in the hippocampus in the HIV+ compared to the HIV- group and was decreased in the MCMD and HAD subgroups compared to HIV+ controls. Notably the ratio of ERalpha-positive reactive astrocytes to total reactive astrocytes increased and significantly correlated with the severity of cognitive impairment following HIV infection. The data suggest that E2 would have the most dramatic effect in reducing HIV transcription early in the disease process when the subpopulation of astrocytes expressing ERalpha is low.
Collapse
Affiliation(s)
- Paula M. Heron
- Department of Physiology, University of Kentucky Medical Center, Lexington, Kentucky 40536-0298
| | - Jadwiga Turchan-Cholewo
- Department of Anatomy and Neurobiology, University of Kentucky Medical Center, Lexington, Kentucky 40536-0298
| | - Annadora J. Bruce-Keller
- Department of Anatomy and Neurobiology, University of Kentucky Medical Center, Lexington, Kentucky 40536-0298
- Current address: Pennington Biomedical Research Center/Louisiana State University, Baton Rouge, Louisiana 70808
| | - Melinda E. Wilson
- Department of Physiology, University of Kentucky Medical Center, Lexington, Kentucky 40536-0298
| |
Collapse
|
76
|
Abstract
The mechanisms of brain ischemic insult include glutamate excitoxicity, calcium toxicity, free radicals, nitric oxide, inflammatory reactions, as well as dysfunctions of endoplasmic reticulum and mitochondrion. These injury cascades are interconnected in complex ways, thus it is hard to compare their pathogenic importances in ischemia models. And the research in cellular and molecular pathways has spurred the studies in potential neuroprotections mainly in pharmacological fields, such as anti-excitotoxic treatment, calcium-channel antagonism, approaches for inhibition of oxidation, inflammation and apoptosis, etc. Besides, other protective interventions including thrombolysis, arteriogenesis, regeneration therapy, and ischemia preconditioning or postconditioning, are also under investigations. Despite the present difficulties, we are quite optimistic towards future clinical applications of neuroprotective agents, by optimizing experimental approaches and clinical trials.
Collapse
|
77
|
Garrett JE, Wellman CL. Chronic stress effects on dendritic morphology in medial prefrontal cortex: sex differences and estrogen dependence. Neuroscience 2009; 162:195-207. [PMID: 19401219 PMCID: PMC2720075 DOI: 10.1016/j.neuroscience.2009.04.057] [Citation(s) in RCA: 164] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2009] [Revised: 04/20/2009] [Accepted: 04/22/2009] [Indexed: 01/07/2023]
Abstract
A growing body of work has documented sex differences in many behavioral, neurochemical, and morphological responses to stress. Chronic stress alters morphology of dendrites in medial prefrontal cortex in male rats. However, potential sex differences in stress-induced morphological changes in medial prefrontal cortex have not been examined. Thus, in Experiment 1 we assessed dendritic morphology in medial prefrontal cortex in male and female rats after chronic stress. Male and female rats underwent either 3 hours of restraint daily for 1 week or were left unhandled except for weighing. On the final day of restraint, all rats were euthanized and brains were stained using a Golgi-Cox procedure. Pyramidal neurons in layer II-III of medial prefrontal cortex were drawn in three dimensions, and morphology of apical and basilar arbors was quantified. In males, stress decreased apical dendritic branch number and length, whereas in females, stress increased apical dendritic length. In Experiment 2, we assessed whether estradiol mediates this stress-induced dendritic hypertrophy in females by assessing the effects of restraint stress on female rats that had received either ovariectomy with or without 17-beta-estradiol replacement or sham ovariectomy. Brains were processed and neurons reconstructed as described in Experiment 1. Both sham-operated and ovariectomized rats with estradiol implants showed stress-induced increases in apical dendritic material, whereas ovariectomy without estradiol replacement prevented the stress-induced increase. Thus, the stress-induced increase in apical dendritic material in females is estradiol-dependent.
Collapse
Affiliation(s)
- J E Garrett
- Department of Psychological and Brain Sciences and Program in Neuroscience, 1101 East 10th Street, Indiana University, Bloomington, IN 47405, USA
| | | |
Collapse
|
78
|
Vanoye-Carlo A, Mendoza-Rodriguez CA, Morales T, Langley E, Cerbón M. Estrogen receptors increased expression during hippocampal neuroprotection in lactating rats. J Steroid Biochem Mol Biol 2009; 116:1-7. [PMID: 19467858 DOI: 10.1016/j.jsbmb.2009.02.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2008] [Revised: 02/10/2009] [Accepted: 02/12/2009] [Indexed: 11/28/2022]
Abstract
Estrogen receptor (ER)-mediated neuroprotection has been demonstrated in both in vitro and in vivo model systems. Two types of estrogen receptors, ERalpha and ERbeta, are the major mediators of the biological functions of estrogens. In the hippocampus, ERbeta is prevalent over ERalpha. Recently, we reported that during the final phase of lactation there is a neuroprotective mechanism in the hippocampus of the adult female rat against neuronal damage induced by systemic kainic acid administration vs. virgin (metestrus) rats. In this study, we assessed differential ER expression and localization in CA1, CA3 and dentate gyrus regions of dorsal hippocampus of metestrus and lactating adult rats at day 19 of lactation, during basal conditions (metestrus and L19, respectively) and 24h after systemic kainate administration. ERs were assessed by western blot and immunohistochemistry. We found a significant increase in the expression of ERs in the hippocampus during lactation as compared with metestrus. ERbeta was significantly increased in the CA1 and CA3 of lactating rats after the kainic acid insult. In addition, we observed a relocalization of ERbeta from the cytoplasm to the nucleus of neuronal cells. Our results suggest that there is a strong correlation between expression of ERs, especially ERbeta, in lactating CA1 and CA3 hippocampus regions in response to kainate administration, and neuroprotection observed during this reproductive period. This may be one of the mechanisms involved in the protection of the maternal brain to ensure offspring survival.
Collapse
Affiliation(s)
- América Vanoye-Carlo
- Departamento de Biología, Facultad de Química, Universidad Nacional Autónoma de México, México, D.F. 04510, Mexico
| | | | | | | | | |
Collapse
|
79
|
Strom JO, Theodorsson A, Theodorsson E. Dose-related neuroprotective versus neurodamaging effects of estrogens in rat cerebral ischemia: a systematic analysis. J Cereb Blood Flow Metab 2009; 29:1359-72. [PMID: 19458604 DOI: 10.1038/jcbfm.2009.66] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Numerous studies of the effects of estrogens for stroke prevention have yielded conflicting results in human and animal studies alike. We present a systematical analysis of study design and methodological differences between 66 studies where estrogens' impact on ischemic brain damage in rat models has been investigated, providing evidence that the differences in results may be explained by high estrogen doses produced by slow-release pellets. These pellets have been used in all studies showing increased neurologic damage because of estrogens. Our data indicate that the increased neurologic damage is related to the pellets' plasma concentration profile with an early, prolonged, supraphysiological peak. Neither the method of inducing the ischemic brain lesions, the choice of variables for measuring outcome, the measured plasma concentrations of estrogens at the time of ischemia nor rat population attributes (sex, strain, age, and diseases) are factors contributing to the discrepancies in results. This suggests that the effects of estrogens for stroke prevention are concentration related with a complex dose-response curve, and underscores the importance of carefully validating the experimental methods used. Future studies of hormone-replacement therapy in women may have to take dosage and administration regimens into account.
Collapse
Affiliation(s)
- Jakob O Strom
- Department of Clinical Chemistry, Institution of Clinical and Experimental Medicine, Linköping University Hospital, Linköping, Sweden
| | | | | |
Collapse
|
80
|
Bethea CL, Reddy AP, Tokuyama Y, Henderson JA, Lima FB. Protective actions of ovarian hormones in the serotonin system of macaques. Front Neuroendocrinol 2009; 30:212-38. [PMID: 19394356 PMCID: PMC2704571 DOI: 10.1016/j.yfrne.2009.04.003] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2009] [Revised: 04/14/2009] [Accepted: 04/15/2009] [Indexed: 12/19/2022]
Abstract
The serotonin neurons of the dorsal and medial raphe nuclei project to all areas of the forebrain and play a key role in mood disorders. Hence, any loss or degeneration of serotonin neurons could have profound ramifications. In a monkey model of surgical menopause with hormone replacement and no neural injury, E and P decreased gene expression in the dorsal raphe nucleus of c-jun n-terminal kinase (JNK1) and kynurenine mono-oxygenase (KMO) that promote cell death. In concert, E and P increased gene expression of superoxide dismutase (SOD1), VEGF, and caspase inhibitory proteins that promote cellular resilience in the dorsal raphe nucleus. Subsequently, we showed that ovarian steroids inhibit pivotal genes in the caspase-dependent and caspase-independent pathways in laser-captured serotonin neurons including apoptosis activating factor (Apaf1), apoptosis-inducing factor (AIF) and second mitochondria-derived activator of caspases (Smac/Diablo). SOD1 was also increased specifically in laser-captured serotonin neurons. Examination of protein expression in the dorsal raphe block revealed that JNK1, phosphoJNK1, AIF and the translocation of AIF from the mitochondria to the nucleus decreased with hormone therapy, whereas pivotal execution proteins in the caspase pathway were unchanged. In addition, cyclins A, B, D1 and E were inhibited, which would prevent re-entry into the cell cycle and catastrophic death. These data indicated that in the absence of gross injury to the midbrain, ovarian steroids inhibit the caspase-independent pathway and cell cycle initiation in serotonin neurons. To determine if these molecular actions prevented cellular vulnerability or death, we examined DNA fragmentation in the dorsal raphe nucleus with the TUNEL assay (terminal deoxynucleotidyl transferase nick end labeling). Ovarian steroids significantly decreased the number of TUNEL-positive cells in the dorsal raphe. Moreover, TUNEL staining prominently colocalized with TPH immunostaining, a marker for serotonin neurons. In summary, ovarian steroids increase the cellular resilience of serotonin neurons and may prevent serotonin neuron death in women facing decades of life after menopause. The survival of serotonin neurons would support cognition and mental health.
Collapse
Affiliation(s)
- Cynthia L Bethea
- Divisions of Reproductive Sciences, Oregon National Primate Research Center, Beaverton, OR 97006, United States.
| | | | | | | | | |
Collapse
|
81
|
|
82
|
Neuroprotection against excitotoxic brain injury in mice after ovarian steroid depletion. Brain Res 2009; 1265:37-46. [PMID: 19236850 DOI: 10.1016/j.brainres.2009.02.023] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2008] [Accepted: 02/10/2009] [Indexed: 11/21/2022]
Abstract
Ovarian steroid hormones influence not only seizure phenomena, but also the neuronal cell death that follows. In the present study, we applied two models of ovarian steroid loss, ovariectomy and chemically-induced ovarian failure, to evaluate kainate-induced seizure activity and the susceptibility of hippocampal neurons to seizure-induced neurodegeneration. Young adult female FVB/NJ mice were ovariectomized with (OVX+E, n=6) or without (OVX, n=8) estrogen replacement. A separate group of females received the ovotoxin, 4-vinylcyclohexene diepoxide (VCD, n=8) to deplete ovarian follicles. Mice underwent kainate-induced status epilepticus and were evaluated for seizure activity (3 h) and delayed hippocampal neuronal injury (7 days). While there were no differences in latency or duration of severe seizures among control, OVX and VCD-treated mice, OVX+E mice exhibited seizures of a significantly longer duration. However, both VCD-induced ovarian failure and OVX led to a dramatic reduction in the extent of excitotoxic cell death, with slightly greater effects observed in VCD-treated mice. Estradiol administration to OVX mice also exerted a significant neuroprotective effect against kainate-induced cell death. These results support and extend earlier findings suggesting that the hormonal milieu may have differential effects on seizure susceptibility that are separate and distinct from those influencing hippocampal neuronal vulnerability. Collectively, these findings highlight the complex interactions among the loss of ovarian steroid hormones, estrogen replacement, seizures, and seizure-induced cell death.
Collapse
|
83
|
VanLandingham JW, Cekic M, Cutler SM, Hoffman SW, Washington ER, Johnson SJ, Miller D, Stein DG. Progesterone and its metabolite allopregnanolone differentially regulate hemostatic proteins after traumatic brain injury. J Cereb Blood Flow Metab 2008; 28:1786-94. [PMID: 18628783 DOI: 10.1038/jcbfm.2008.73] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Our laboratory has shown in numerous experiments that the neurosteroids progesterone (PROG) and allopregnanolone (ALLO) improve molecular and functional outcomes after traumatic brain injury (TBI). As coagulopathy is an important contributor to the secondary destruction of nervous tissue, we hypothesized that PROG and ALLO administration may also have a beneficial effect on coagulation protein expression after TBI. Adult male Sprague-Dawley rats were given bilateral contusions of the medial frontal cortex followed by treatments with PROG (16 mg/kg), ALLO (8 mg/kg), or vehicle (22.5% hydroxypropyl-beta-cyclodextrin). Controls received no injury or injections. Progesterone generally maintained procoagulant (thrombin, fibrinogen, and coagulation factor XIII), whereas ALLO increased anticoagulant protein expression (tissue-type plasminogen activator, tPA). In addition, PROG significantly increased the ratio of tPA bound to neuroserpin, a serine protease inhibitor that can reduce the activity of tPA. Our findings suggest that in a model of TBI, where blood loss may exacerbate injury, it may be preferable to treat patients with PROG, whereas it might be more appropriate to use ALLO as a treatment for thrombotic stroke, where a reduction in coagulation would be more beneficial.
Collapse
Affiliation(s)
- Jacob W VanLandingham
- Department of Emergency Medicine, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | | | | | | | | | | | | | | |
Collapse
|
84
|
Jia J, Guan D, Zhu W, Alkayed NJ, Wang MM, Hua Z, Xu Y. Estrogen inhibits Fas-mediated apoptosis in experimental stroke. Exp Neurol 2008; 215:48-52. [PMID: 18950622 DOI: 10.1016/j.expneurol.2008.09.015] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2008] [Revised: 09/05/2008] [Accepted: 09/11/2008] [Indexed: 12/11/2022]
Abstract
Estrogen is protective in experimental cerebral ischemia, yet the mechanism remains unclear. Fas-mediated apoptosis has been shown to be induced after cerebral ischemia and significantly contribute to ischemic brain damage. In this study, we tested if estrogen is protective against cerebral ischemia by suppressing Fas-mediated apoptosis. 17Beta-estradiol-treated and untreated ovariectomized (OVX) female mice were subjected to 2 h middle cerebral artery occlusion (MCAO). Expression of Fas and Fas-associated death domain (FADD) were measured at 3, 6 and 12 h of reperfusion by RT-PCR and Western blot, respectively. Post-ischemic activities of caspase-8 and -3 activities, the two downstream effectors of Fas-induced apoptosis, were also assayed at same time points by ELISA. Finally, Fas antibody-induced cell death in primary cortical neurons was assayed by fluorescence activated cell sorter (FACS) in the presence and absence of estradiol. Our data showed that estradiol-treated OVX female mice sustained smaller infarct compared to untreated OVX mice. Ischemia upregulated Fas and FADD expression, and increased caspase-8 and -3 activities in OVX female mouse cortex, which were significantly attenuated by estradiol. Estradiol also significantly inhibited Fas antibody-induced neuronal cell apoptosis. Our data suggests that inhibition of ischemia-induced Fas-mediated apoptosis is an important mechanism of neuroprotection by estrogen in cerebral ischemia.
Collapse
Affiliation(s)
- Jia Jia
- Department of Neurology, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, 210008, PR China
| | | | | | | | | | | | | |
Collapse
|
85
|
Abstract
Anesthesiologists are frequently confronted with patients who are at risk for neurological complications due to perioperative stroke or prior traumatic brain injury. In this review, we address the growing and fascinating body of data that suggests gender and sex steroids influence the pathophysiology of injury and outcome for these patients. Cerebral ischemia, traumatic brain injury, and epilepsy are reviewed in the context of potential sex differences in mechanisms and outcomes of brain injury and the role of estrogen, progesterone, and androgens in shaping these processes. Lastly, implications for current and future perioperative and intensive care are identified.
Collapse
Affiliation(s)
- Kamila Vagnerova
- Department of Anesthesiology and Peri-Operative Medicine, Oregon Health and Science University, Portland, Oregon 97239, USA
| | | | | |
Collapse
|
86
|
Abstract
The ability to reduce brain injury before, during or after an ischaemic injury, irrespective of the cause, remains an exciting prospect. In this article, we will discuss some of the current research behind cerebral protection, which will include the use of anaesthetic agents, as well as therapies targeted specifically at the complex cascades following brain injury.
Collapse
Affiliation(s)
- Jane Sturgess
- Cambridge University Hospitals Foundation Trust, Hills Road, Cambridge CB2 2QQ, UK
| | | |
Collapse
|
87
|
Antiseizure effects of 5*-androstane-3*,7beta-diol may be independent of actions at estrogen receptor beta. Epilepsy Behav 2008; 13:32-5. [PMID: 18455477 DOI: 10.1016/j.yebeh.2008.03.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2007] [Revised: 03/25/2008] [Accepted: 03/26/2008] [Indexed: 11/23/2022]
Abstract
Testosterone (T), the principal androgen secreted by the testes, can have antiseizure effects; however, the mechanism(s) underlying this action is not well understood. T is metabolized to dihydrotestosterone (DHT) by the enzyme 5*-reductase. DHT is then converted to 5*-androstane-3*,17beta-diol (3*-diol) by the enzyme 3*-hydroxysteroid dehydrogenase. T and DHT bind with high affinity to intracellular androgen receptors; however, 3*-diol does not. The mnemonic effects of 3*-diol are mediated in part through the beta isoform of estrogen receptors (ERbeta) in the hippocampus. As such, we investigated whether 3*-diol has antiseizure effects in mice that require action at ERbeta. 3*-Diol (2 mg/kg subcutaneously) was administered to wild-type C57/B6 mice and heterozygous and homozygous ERbeta knockout (betaERKO) mice 1 hour prior to administration of pentylenetetrazol (PTZ; 85 mg/kg intraperitoneally). Mice administered 3*-diol had significantly longer latencies to clonic seizure and death and lower seizure scores than did mice administered vehicle. This pattern of effects was observed in wild-type or betaERKO mice. Thus, for these mice, the antiseizure effects of 3*-diol for the chemoconvulsant PTZ occur independent of actions at ERbeta.
Collapse
|
88
|
Foster TC, Rani A, Kumar A, Cui L, Semple-Rowland SL. Viral vector-mediated delivery of estrogen receptor-alpha to the hippocampus improves spatial learning in estrogen receptor-alpha knockout mice. Mol Ther 2008; 16:1587-1593. [PMID: 18594506 DOI: 10.1038/mt.2008.140] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2007] [Accepted: 03/06/2008] [Indexed: 01/01/2023] Open
Abstract
Estrogen, which influences both classical genomic and rapid membrane-associated signaling cascades, has been implicated in the regulation of hippocampal function, including spatial learning. Gene mutation studies suggest that estrogen effects are mediated by estrogen receptor-alpha (ER-alpha); however, because gonadal steroids influence the organization of the hippocampus during development, it has been difficult to distinguish developmental effects from those specific to adults. In this study we show that lentiviral delivery of the gene encoding ER-alpha to the hippocampus of adult ER-alpha-knockout (ER-alphaKO) mice restores hippocampal responsiveness to estrogen and rescues spatial learning. We propose that constitutive estrogen receptor activity is important for maintaining hippocampus-dependent memory function in adults.
Collapse
Affiliation(s)
- Thomas C Foster
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, Florida 32610-0244, USA.
| | | | | | | | | |
Collapse
|
89
|
Lang JT, McCullough LD. Pathways to ischemic neuronal cell death: are sex differences relevant? J Transl Med 2008; 6:33. [PMID: 18573200 PMCID: PMC2459157 DOI: 10.1186/1479-5876-6-33] [Citation(s) in RCA: 148] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2008] [Accepted: 06/23/2008] [Indexed: 11/24/2022] Open
Abstract
We have known for some time that the epidemiology of human stroke is sexually dimorphic until late in life, well beyond the years of reproductive senescence and menopause. Now, a new concept is emerging: the mechanisms and outcome of cerebral ischemic injury are influenced strongly by biological sex as well as the availability of sex steroids to the brain. The principal mammalian estrogen (17 β estradiol or E2) is neuroprotective in many types of brain injury and has been the major focus of investigation over the past several decades. However, it is becoming increasingly clear that although hormones are a major contributor to sex-specific outcomes, they do not fully account for sex-specific responses to cerebral ischemia. The purpose of this review is to highlight recent studies in cell culture and animal models that suggest that genetic sex determines experimental stroke outcome and that divergent cell death pathways are activated after an ischemic insult. These sex differences need to be identified if we are to develop efficacious neuroprotective agents for use in stroke patients.
Collapse
Affiliation(s)
- Jesse T Lang
- Department of Neurology, The University of Connecticut Health Center, Farmington, CT, USA.
| | | |
Collapse
|
90
|
Wahl AS, Buchthal B, Rode F, Bomholt SF, Freitag HE, Hardingham GE, Rønn LCB, Bading H. Hypoxic/ischemic conditions induce expression of the putative pro-death gene Clca1 via activation of extrasynaptic N-methyl-D-aspartate receptors. Neuroscience 2008; 158:344-52. [PMID: 18616988 DOI: 10.1016/j.neuroscience.2008.06.018] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2007] [Revised: 03/13/2008] [Accepted: 06/08/2008] [Indexed: 10/24/2022]
Abstract
The stimulation of extrasynaptic N-methyl-D-aspartate (NMDA) receptors triggers cell death pathways and has been suggested to play a key role in cell degeneration and neuron loss associated with glutamate-induced excitotoxicity. In contrast, synaptic NMDA receptors promote neuronal survival. One mechanism through which extrasynaptic NMDA receptors damage neurons may involve Clca1, which encodes a putative calcium-activated chloride channel. Here we show that Clca1 expression is induced in cultured rat hippocampal neurons exposed to oxygen/glucose-free media; this induction is mediated by a signaling pathway activated by extrasynaptic NMDA receptors. Clca1 mRNA levels also increased in the gerbil hippocampus following a transient forebrain ischemia caused by bilateral carotid occlusion. Microelectrode array recordings revealed that oxygen-glucose deprivation enhances hippocampal network firing rates, which induces c-fos transcription through a signaling pathway that, in contrast to Clca1, is activated by synaptic but not extrasynaptic NMDA receptors. Thus, conditions of low oxygen/glucose lead to the activation of both extrasynaptic and synaptic NMDA receptors that regulate distinct target genes. Clca1 may be part of the genomic death program triggered by extrasynaptic NMDA receptors; it could be a marker for ischemic brain damage and a possible target for therapeutic interventions.
Collapse
Affiliation(s)
- A-S Wahl
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), University of Heidelberg, Im Neuenheimer Feld 364, 69120 Heidelberg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
91
|
Hendricks JL, Chikar JA, Crumling MA, Raphael Y, Martin DC. Localized cell and drug delivery for auditory prostheses. Hear Res 2008; 242:117-31. [PMID: 18573323 DOI: 10.1016/j.heares.2008.06.003] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2007] [Revised: 05/09/2008] [Accepted: 06/02/2008] [Indexed: 12/20/2022]
Abstract
Localized cell and drug delivery to the cochlea and central auditory pathway can improve the safety and performance of implanted auditory prostheses (APs). While generally successful, these devices have a number of limitations and adverse effects including limited tonal and dynamic ranges, channel interactions, unwanted stimulation of non-auditory nerves, immune rejection, and infections including meningitis. Many of these limitations are associated with the tissue reactions to implanted auditory prosthetic devices and the gradual degeneration of the auditory system following deafness. Strategies to reduce the insertion trauma, degeneration of target neurons, fibrous and bony tissue encapsulation, and immune activation can improve the viability of tissue required for AP function as well as improve the resolution of stimulation for reduced channel interaction and improved place-pitch and level discrimination. Many pharmaceutical compounds have been identified that promote the viability of auditory tissue and prevent inflammation and infection. Cell delivery and gene therapy have provided promising results for treating hearing loss and reversing degeneration. Currently, many clinical and experimental methods can produce extremely localized and sustained drug delivery to address AP limitations. These methods provide better control over drug concentrations while eliminating the adverse effects of systemic delivery. Many of these drug delivery techniques can be integrated into modern auditory prosthetic devices to optimize the tissue response to the implanted device and reduce the risk of infection or rejection. Together, these methods and pharmaceutical agents can be used to optimize the tissue-device interface for improved AP safety and effectiveness.
Collapse
Affiliation(s)
- Jeffrey L Hendricks
- Department of Biomedical Engineering, The University of Michigan, 1107 Gerstacker Building, 2200 Bonisteel Boulevard, Ann Arbor, MI 48109-2099, USA.
| | | | | | | | | |
Collapse
|
92
|
Sun M, Xu C. Neuroprotective mechanism of taurine due to up-regulating calpastatin and down-regulating calpain and caspase-3 during focal cerebral ischemia. Cell Mol Neurobiol 2008; 28:593-611. [PMID: 17712625 DOI: 10.1007/s10571-007-9183-8] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2007] [Accepted: 07/27/2007] [Indexed: 10/22/2022]
Abstract
AIMS Taurine as an endogenous substance possesses a number of cytoprotective properties. In the study, we have evaluated the neuroprotective effect of taurine and investigated whether taurine exerted neuroprotection through affecting calpain/calpastatin or caspase-3 actions during focal cerebral ischemia, since calpain and caspase-3 play central roles in ischemic neuronal death. METHODS Male Sprague-Dawley rats were subjected to 2 h of middle cerebral artery occlusion (MCAo), and 22 h of reperfusion. Taurine was administrated intravenously 1 h after MCAo. The dose-responses of taurine to MCAo were determined. Next, the effects of taurine on the activities of calpain, calpastatin and caspase-3, the levels of calpastatin, microtubule-associated protein-2 (MAP-2) and alphaII-spectrin, and the apoptotic cell death in penumbra were evaluated. RESULTS Taurine reduced neurological deficits and decreased the infarct volume 24 h after MCAo in a dose-dependent manner. Treatment with 50 mg/kg of taurine significantly increased the calpastatin protein levels and activities, and markedly reduced the m-calpain and caspase-3 activities in penumbra 24 h after MCAo, however, it had no significant effect on mu-calpain activity. Moreover, taurine significantly increased the MAP-2 and alphaII-spectrin protein levels, and markedly reduced the ischemia-induced TUNEL staining positive score within penumbra 24 h after MCAo. CONCLUSIONS Our data demonstrate the dose-dependent neuroprotection of taurine against transient focal cerebral ischemia, and suggest that one of protective mechanisms of taurine against ischemia may be blocking the m-calpain and caspase-3-mediated apoptotic cell death pathways.
Collapse
Affiliation(s)
- Ming Sun
- Department of Neurochemistry, Beijing Neurosurgical Institute, 6 Tiantan Xi Li, 100050 Beijing, PR China
| | | |
Collapse
|
93
|
Bode FJ, Stephan M, Suhling H, Pabst R, Straub RH, Raber KA, Bonin M, Nguyen HP, Riess O, Bauer A, Sjoberg C, Petersén A, von Hörsten S. Sex differences in a transgenic rat model of Huntington's disease: decreased 17beta-estradiol levels correlate with reduced numbers of DARPP32+ neurons in males. Hum Mol Genet 2008; 17:2595-609. [PMID: 18502785 DOI: 10.1093/hmg/ddn159] [Citation(s) in RCA: 97] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Recent clinical studies have highlighted that female sex hormones represent potential neuroprotective mediators against damage caused by acute and chronic brain diseases. This evidence has been confirmed by experimental studies documenting the protective role of female sex hormones both in vitro and in vivo, although these studies did not specifically focus on Huntington's disease (HD). We therefore investigated the onset and course of HD in female and male transgenic (tg) HD (CAG(n51)) and control rats across age and focused on three aspects: (i) behavioral and physiological alterations (energy expenditure, home-cage activity, emotional disturbance and motor dysfunction), (ii) morphological markers (numbers and characteristics of striatal DARPP32(+) medium-sized spiny neurons (MSNs) and dopamine receptor autoradiography) and (iii) peripheral sex hormone levels as well as striatal estrogen receptor expression. Independent of their sex, tgHD rats exhibited increased levels of food intake, elevated home-cage activity scores and anxiolytic-like behavior, whereas only males showed an impairment of motor function. In line with the latter finding, loss and atrophy of DARPP32(+) MSNs were apparent only in male tgHD rats. This result was associated with a decreased striatal dopamine D1 receptor density and lower plasma levels of 17beta-estradiol at the age of 14 months. As DARPP32(+) MSNs expressed both alpha- and beta-estrogen receptors and showed a correlation between cell numbers and 17beta-estradiol levels, our findings suggest sex-related differences in the HD phenotype pointing to a substantial neuroprotective effect of sex hormones and opening new perspectives on the therapy of HD.
Collapse
Affiliation(s)
- Felix J Bode
- Institute of Functional and Applied Anatomy, Medical School of Hannover, 30625 Hannover, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
94
|
Sengelaub DR, Forger NG. The spinal nucleus of the bulbocavernosus: firsts in androgen-dependent neural sex differences. Horm Behav 2008; 53:596-612. [PMID: 18191128 PMCID: PMC2423220 DOI: 10.1016/j.yhbeh.2007.11.008] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2007] [Revised: 11/01/2007] [Accepted: 11/05/2007] [Indexed: 11/30/2022]
Abstract
Cell number in the spinal nucleus of the bulbocavernosus (SNB) of rats was the first neural sex difference shown to differentiate under the control of androgens, acting via classical intracellular androgen receptors. SNB motoneurons reside in the lumbar spinal cord and innervate striated muscles involved in copulation, including the bulbocavernosus (BC) and levator ani (LA). SNB cells are much larger and more numerous in males than in females, and the BC/LA target muscles are reduced or absent in females. The relative simplicity of this neuromuscular system has allowed for considerable progress in pinpointing sites of hormone action, and identifying the cellular bases for androgenic effects. It is now clear that androgens act at virtually every level of the SNB system, in development and throughout adult life. In this review we focus on effects of androgens on developmental cell death of SNB motoneurons and BC/LA muscles; the establishment and maintenance of SNB motoneuron soma size and dendritic length; BC/LA muscle morphology and physiology; and behaviors controlled by the SNB system. We also describe new data on neurotherapeutic effects of androgens on SNB motoneurons after injury in adulthood.
Collapse
Affiliation(s)
- Dale R Sengelaub
- Program in Neuroscience, Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN 47405, USA.
| | | |
Collapse
|
95
|
Estradiol-17beta protects against hypoxia-induced hepatocyte injury through ER-mediated upregulation of Bcl-2 as well as ER-independent antioxidant effects. Cell Res 2008; 18:491-9. [PMID: 18379592 DOI: 10.1038/cr.2008.42] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Although many previous studies have suggested that estrogen functions as a cytoprotective agent under oxidative stress conditions, the underlying mechanism by which this effect is exerted remains to be elucidated. This study assessed the effects of estradiol-17beta (E(2)) (10(-8) M) on hypoxia-induced cell injury and its related signaling in primary cultured chicken hepatocytes. Hypoxic conditions were found to augment the level of DNA damage and to reduce cell viability and the level of [(3)H]-thymidine incorporation, and these phenomena were prevented through treatment with E(2). Hypoxia also increased caspase-3 expression, but showed no evidence of an influence on the expression of Bcl-2. However, E(2) induced an increase in the level of Bcl-2 expression under hypoxic conditions and reduced the level of caspase-3 expression. The effects of E(2) on Bcl-2 and caspase expression were blocked by ICI 182780 (E(2) receptor (ER) antagonist, 10(-7) M). In addition, hypoxia resulted in an increase in the intracellular reactive oxygen species (ROS) generated. These effects were blocked by E(2), but not by E(2)-BSA and ICI 182780. Hypoxia also activated p38 mitogen-activated protein kinase (MAPK), c-JUN N-terminal kinase/stress-activated protein kinase (JNK/SAPK) and nuclear factor-kappaB (NF-kappaB). These effects were blocked by E(2), but not by ICI 182780. The inhibition of p38 MAPK and JNK/SAPK blocked NF-kappaB activation. In conclusion, E(2) was found to protect against hypoxia-induced cell injury in chicken hepatocytes through ER-mediated upregulation of Bcl-2 expression and through reducing the activity of ROS-dependent p38 MAPK, JNK/SAPK and NF-kappaB.
Collapse
|
96
|
Tokuyama Y, Reddy AP, Bethea CL. Neuroprotective actions of ovarian hormones without insult in the raphe region of rhesus macaques. Neuroscience 2008; 154:720-31. [PMID: 18486349 DOI: 10.1016/j.neuroscience.2008.03.056] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2007] [Revised: 02/11/2008] [Accepted: 03/12/2008] [Indexed: 11/17/2022]
Abstract
Using a nonhuman primate model of surgical menopause, our laboratory has shown that ovarian hormone treatment (HT) improves 5-HT neural function in the dorsal raphe nucleus (DRN). We further hypothesize that HT may increase 5-HT neuronal resilience. Recent data from microarray analysis indicated that HT regulates gene expression in pathways that lead to apoptosis. In this study, we questioned whether HT alters protein expression in caspase-dependent and independent pathways. Ovariectomized monkeys received Silastic implants containing placebo (empty), estrogen (E) or E+ progesterone (P). A small block of the midbrain containing the DRN was dissected and subjected to subcellular fractionation, yielding cytosolic, nuclear and mitochondrial fractions (n=4/group). The pro-apoptotic protein, c-jun n-terminal kinase (JNK1) and its phosphorylation were decreased by E+P treatment in the cytosolic fraction. Downstream of JNK are proteins in the caspase-dependent and -independent pathways. First, in the caspase-dependent pathway, cytoplasmic and mitochondrial fractions were immunoblotted for Bcl-2 family members, cytochrome c, Apaf1 and XIAP. However, the expression of these proteins did not differ among treatments. Pro-caspase 3 was decreased by E+P, but there was no evidence of active caspase in any group. Then, we examined the involvement of a protein in the caspase-independent pathway, called apoptosis-inducing factor (AIF). AIF mRNA (n=3/group) and AIF mitochondrial protein tended to decrease with hormone treatment. However, AIF protein in the nuclear fraction in E+P treated monkeys was significantly reduced. This indicates that HT is reducing the translocation of AIF from mitochondria to nucleus, thus inhibiting AIF-mediated apoptosis. AIF was immunocytochemically localized to large 5-HT-like neurons of the dorsal raphe. These data suggest that in the absence of global trauma or ischemia, HT may act through the caspase-independent pathway to promote neuroprotection in the 5-HT system.
Collapse
Affiliation(s)
- Y Tokuyama
- Division of Reproductive Sciences, Oregon National Primate Research Center, Beaverton, OR 97006, USA
| | | | | |
Collapse
|
97
|
Morissette M, Le Saux M, D'Astous M, Jourdain S, Al Sweidi S, Morin N, Estrada-Camarena E, Mendez P, Garcia-Segura LM, Di Paolo T. Contribution of estrogen receptors alpha and beta to the effects of estradiol in the brain. J Steroid Biochem Mol Biol 2008; 108:327-38. [PMID: 17936613 DOI: 10.1016/j.jsbmb.2007.09.011] [Citation(s) in RCA: 142] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Clinical and experimental studies show a modulatory role of estrogens in the brain and suggest their beneficial action in mental and neurodegenerative diseases. The estrogen receptors ERalpha and ERbeta are present in the brain and their targeting could bring selectivity and reduced risk of cancer. Implication of ERs in the effect of estradiol on dopamine, opiate and glutamate neurotransmission is reviewed. The ERalpha agonist, PPT, is shown as estradiol to modulate hippocampal NMDA receptors and AMPA receptors in cortex and striatum of ovariectomized rats whereas the ERbeta agonist DPN is inactive. Striatal DPN activity suggests implication of ERbeta in estradiol modulation of D2 receptors and transporters in ovariectomized rats and is supported by the lack of effect of estradiol in ERbeta knockout (ERKObeta) mice. Both ERalpha and ERbeta agonists modulate striatal preproenkephalin (PPE) gene expression in ovariectomized rats. In male mice PPT protects against MPTP toxicity to striatal dopamine; this implicates Akt/GSK3beta signaling and the apoptotic regulators Bcl2 and Bad. This suggests a role for ERalpha in striatal dopamine neuroprotection. ERKOalpha mice are more susceptible to MPTP toxicity and not protected by estradiol; differences in ERKObeta mice are subtler. These results suggest therapeutic potential for the brain of ER specific agonists.
Collapse
Affiliation(s)
- M Morissette
- Molecular Endocrinology and Oncology Research Center, Medical Center and Faculty of Pharmacy, Laval University, 2705 Laurier Boulevard, Sainte-Foy, Québec, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
98
|
Stein DG, Wright DW, Kellermann AL. Does Progesterone Have Neuroprotective Properties? Ann Emerg Med 2008; 51:164-72. [PMID: 17588708 DOI: 10.1016/j.annemergmed.2007.05.001] [Citation(s) in RCA: 109] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2006] [Revised: 04/26/2007] [Accepted: 05/01/2007] [Indexed: 11/23/2022]
Abstract
In this article, we review published preclinical and epidemiologic studies that examine progesterone's role in the central nervous system. Its effects on the reproductive and endocrine systems are well known, but a large and growing body of evidence, including a recently published pilot clinical trial, indicates that the hormone also exerts neuroprotective effects on the central nervous system. We now know that it is produced in the brain, for the brain, by neurons and glial cells in the central and peripheral nervous system of both male and female individuals. Laboratories around the world have reported that administering relatively large doses of progesterone during the first few hours to days after injury significantly limits central nervous system damage, reduces loss of neural tissue, and improves functional recovery. Although the research published to date has focused primarily on progesterone's effects on blunt traumatic brain injury, there is evidence that the hormone affords protection from several forms of acute central nervous system injury, including penetrating brain trauma, stroke, anoxic brain injury, and spinal cord injury. Progesterone appears to exert its protective effects by protecting or rebuilding the blood-brain barrier, decreasing development of cerebral edema, down-regulating the inflammatory cascade, and limiting cellular necrosis and apoptosis. All are plausible mechanisms of neuroprotection.
Collapse
Affiliation(s)
- Donald G Stein
- Brain Research Laboratory, Department of Emergency Medicine, School of Medicine, Emory University, Atlanta, GA 30322, USA
| | | | | |
Collapse
|
99
|
Vanoye-Carlo A, Morales T, Ramos E, Mendoza-Rodríguez A, Cerbón M. Neuroprotective effects of lactation against kainic acid treatment in the dorsal hippocampus of the rat. Horm Behav 2008; 53:112-23. [PMID: 17963758 DOI: 10.1016/j.yhbeh.2007.09.004] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2007] [Revised: 08/31/2007] [Accepted: 09/04/2007] [Indexed: 01/20/2023]
Abstract
Marked hippocampal changes in response to excitatory amino acid agonists occur during pregnancy (e.g. decreased frequency in spontaneous recurrent seizures in rats with KA lesions of the hippocampus) and lactation (e.g. reduced c-Fos expression in response to N-methyl-d,l-aspartic acid but not to kainic acid). In this study, the possibility that lactation protects against the excitotoxic damage induced by KA in hippocampal areas was explored. We compared cell damage induced 24 h after a single systemic administration of KA (5 or 7.5 mg/kg bw) in regions CA1, CA3, and CA4 of the dorsal hippocampus of rats in the final week of lactation to that in diestrus phase. To determine cellular damage in a rostro-caudal segment of the dorsal hippocampus, we used NISSL and Fluorojade staining, immunohistochemistry for active caspase-3 and TUNEL, and we observed that the KA treatment provoked a significant loss of neurons in diestrus rats, principally in the pyramidal cells of CA1 region. In contrast, in lactating rats, pyramidal neurons from CA1, CA3, and CA4 in the dorsal hippocampus were significantly protected against KA-induced neuronal damage, indicating that lactation may be a natural model of neuroprotection.
Collapse
Affiliation(s)
- América Vanoye-Carlo
- Departamento de Biología, Facultad de Química, Universidad Nacional Autónoma de México, México, D.F., 04510, Mexico
| | | | | | | | | |
Collapse
|
100
|
Lynch JL, Alley JF, Wellman L, Beitz AJ. Decreased spinal cord opioid receptor mRNA expression and antinociception in a Theiler's murine encephalomyelitis virus model of multiple sclerosis. Brain Res 2007; 1191:180-91. [PMID: 18096140 DOI: 10.1016/j.brainres.2007.11.034] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2007] [Revised: 11/15/2007] [Accepted: 11/17/2007] [Indexed: 01/24/2023]
Abstract
Multiple sclerosis patients typically experience increased pain that is relatively insensitive to opiate treatment. The mechanistic basis for this increased nociception is currently poorly understood. In the present study, we utilized the Theiler's murine encephalomyelitis virus (TMEV) model of MS to examine possible changes in spinal cord opioid receptor mRNA over the course of disease progression. TMEV infection led to significantly decreased mu, delta and kappa opioid receptor mRNA expression as analyzed by quantitative real-time PCR in both male and female mice at days 90, 150 and 180 post-infection (PI). Since opioid receptor mRNA expression decreased in TMEV mice, we examined whether opiate analgesia is also altered. TMEV infected female mice had significantly decreased opiate analgesia in thermal nociceptive tests beginning at day 90 PI, while TMEV-infected male mice did not display significantly decreased opiate analgesia until day 120 PI. The novel finding that opioid receptor expression is significantly decreased in the spinal cord of TMEV mice could explain the increased nociception and loss of opiate analgesia observed in both TMEV mice and multiple sclerosis patients.
Collapse
Affiliation(s)
- Jessica L Lynch
- Department of Veterinary and Biomedical Sciences, University of Minnesota, 1988 Fitch Avenue, St. Paul, MN 55108, USA
| | | | | | | |
Collapse
|