1
|
Ström JO, Ingberg E. Impact of methodology on estrogens' effects on cerebral ischemia in rats: an updated meta-analysis. BMC Neurosci 2014; 15:22. [PMID: 24495535 PMCID: PMC3975994 DOI: 10.1186/1471-2202-15-22] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Accepted: 01/29/2014] [Indexed: 12/15/2022] Open
Abstract
Background Although most animal stroke studies have demonstrated potent neuroprotective effects of estrogens, there are a number of articles reporting the opposite. In 2009, we made the case that this dichotomy was related to administered estrogen dose. Several other suggestions for the discordant results have also been propagated, including the age of the experimental animals and the length of hypoestrogenicity prior to estrogen administration. These two suggestions have gained much popularity, probably because of their kinship with the window of opportunity hypothesis, which is commonly used to explain the analogous dichotomy among human studies. We were therefore encouraged to perform an updated meta-analysis, and to improve it by including all relevant variables in a large multiple regression model, where the impact of confounders could be controlled for. Results The multiple regression model revealed an indisputable impact of estrogen administration mode on the effects of estrogens in ischemic stroke. Subcutaneous slow-release pellets differed from the injection and silastic capsule treatments in terms of impact of estrogens on ischemic stroke, showing that the first mentioned were more prone to render estrogens damaging. Neither the use of elderly animals nor the adoption of longer wash-out periods influenced estrogens’ effects on experimental ischemic stroke in rats. Conclusions We conclude that the discordant results regarding estrogens’ effects in rat models of ischemic stroke are a consequence of differences in estrogen administration modes. These results are not only of importance for the ongoing debate regarding menopausal hormone therapy, but also have an important bearing on experimental stroke methodology and the apparent translational roadblock for suggested stroke interventions.
Collapse
Affiliation(s)
- Jakob O Ström
- Vårdvetenskapligt Forskningscentrum/Centre for Health Sciences, Örebro University Hospital, County Council of Örebro, Örebro SE-703 62, Sweden.
| | | |
Collapse
|
2
|
Strom JO, Ingberg E, Theodorsson E, Theodorsson A. Effects of high and low 17β-estradiol doses on focal cerebral ischemia: negative results. Sci Rep 2013; 3:3111. [PMID: 24177749 PMCID: PMC6505964 DOI: 10.1038/srep03111] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Accepted: 10/11/2013] [Indexed: 01/11/2023] Open
Abstract
The reasons why some animal studies indicate that estrogens increase focal cerebral ischemic damage while others show estrogen-induced neuroprotection has hitherto not been fully elucidated. Recent evidence indicates that discrepancies in hormone administration paradigms, resulting in highly different serum hormone concentrations, may account for the dichotomy. The current study aimed to test this hypothesis. Sixty ovariectomized female rats were randomized into three groups differing in 17β-estradiol regimens, and transient focal cerebral ischemia was subsequently induced. All animals were subjected to a small functional testing battery, and three days after MCAo they were sacrificed for infarct size assessment. Infarct sizes did not differ between groups, however clear discrepancies were seen in body weight and feeding behavior. In comparison to sham-operated animals, ovariectomized rats rapidly increased in body weight, whereas the opposite was seen in rats receiving 17beta-estradiol. The weight gain in the ovariectomized rats was paralleled by an increased food intake.
Collapse
Affiliation(s)
- Jakob O Strom
- Clinical Chemistry, Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University, Department of Clinical Chemistry, County Council of Östergötland, Linköping, Sweden
| | | | | | | |
Collapse
|
3
|
Inagaki T, Etgen AM. Neuroprotective action of acute estrogens: animal models of brain ischemia and clinical implications. Steroids 2013; 78:597-606. [PMID: 23385013 PMCID: PMC3733348 DOI: 10.1016/j.steroids.2012.12.015] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2012] [Revised: 12/20/2012] [Accepted: 12/28/2012] [Indexed: 10/27/2022]
Abstract
The ovarian hormone 17β-estradiol (E2) exerts profound neuroprotective actions against ischemia-induced brain damage in rodent models of global and focal ischemia. This review focuses on the neuroprotective efficacy of post-ischemic administration of E2 and non-feminizing estrogen analogs in the aging brain, with an emphasis on studies in animals subjected to a long-term loss of circulating E2. Clinical findings from the Women's Health Initiative study as well as data from animal studies that used long-term, physiological levels of E2 treatment are discussed in this context. We summarize major published findings that highlight the effective doses and timing of E2 treatment relative to onset of ischemia. We then discuss recent findings from our laboratory showing that under some conditions the aging hippocampus remains responsive to E2 and some neuroprotective non-feminizing estrogen analogs even after prolonged periods of hormone withdrawal. Possible membrane-initiated signaling mechanisms that may underlie the neuroprotective actions of acutely administered E2 are also discussed. Based on these findings, we suggest that post-ischemic treatment with high doses of E2 or certain non-feminizing estrogen analogs may have great therapeutic potential for treatment of brain damage and neurodegeneration associated with ischemia.
Collapse
Affiliation(s)
- Tomoko Inagaki
- Dominick P. Purpura Dept. of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, United States.
| | | |
Collapse
|
4
|
Sohrabji F, Selvamani A, Balden R. Revisiting the timing hypothesis: biomarkers that define the therapeutic window of estrogen for stroke. Horm Behav 2013; 63:222-30. [PMID: 22728278 PMCID: PMC3483414 DOI: 10.1016/j.yhbeh.2012.06.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2012] [Revised: 06/11/2012] [Accepted: 06/12/2012] [Indexed: 02/06/2023]
Abstract
Significantly extended life expectancy coupled with contemporary sedentary lifestyles and poor nutrition has created a global epidemic of cardiovascular disease and stroke. For women, this issue is complicated by the discrepant outcomes of hormone therapy (HT) for stroke incidence and severity as well as the therapeutic complications for stroke associated with advancing age. Here we propose that the impact of estrogen therapy cannot be considered in isolation, but should include age-related changes in endocrine, immune, and nucleic acid mediators that collaborate with estrogen to produce neuroprotective effects commonly seen in younger, healthier demographics. Due to their role as modulators of ischemic cell death, the post-stroke inflammatory response, and neuronal survival and regeneration, this review proposes that Insulin-like Growth Factor (IGF)-1, Vitamin D, and discrete members of the family of non-coding RNA peptides called microRNAs (miRNAs) may be crucial biochemical markers that help determine the neuroprotective "window" of HT. Specifically, IGF-1 confers neuroprotection in concert with, and independently of, estrogen and failure of the insulin/IGF-1 axis is associated with metabolic disturbances that increase the risk for stroke. Vitamin D and miRNAs regulate and complement IGF-1 mediated function and neuroprotective efficacy via modulation of IGF-1 availability and neural stem cell and immune cell proliferation, differentiation and secretions. Together, age-related decline of these factors differentially affects stroke risk, severity, and outcome, and may provide a novel therapeutic adjunct to traditional HT practices.
Collapse
Affiliation(s)
- Farida Sohrabji
- Department of Neuroscience and Experimental Therapeutics, Women's Health in Neuroscience Program, Texas A&M Health Science Center, College Station, TX 77843-1114, USA.
| | | | | |
Collapse
|
5
|
Canal Castro C, Pagnussat AS, Orlandi L, Worm P, Moura N, Etgen AM, Alexandre Netto C. Coumestrol has neuroprotective effects before and after global cerebral ischemia in female rats. Brain Res 2012; 1474:82-90. [DOI: 10.1016/j.brainres.2012.07.025] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2012] [Revised: 06/26/2012] [Accepted: 07/12/2012] [Indexed: 01/29/2023]
|
6
|
Pereno GL, Balaszczuk V, Beltramino CA. Effect of sex differences and gonadal hormones on kainic acid-induced neurodegeneration in the bed nucleus of the stria terminalis of the rat. ACTA ACUST UNITED AC 2012; 64:283-9. [DOI: 10.1016/j.etp.2010.08.020] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2010] [Revised: 08/29/2010] [Accepted: 08/29/2010] [Indexed: 12/12/2022]
|
7
|
Li M, Zhang Z, Sun W, Koehler RC, Huang J. 17β-estradiol attenuates breakdown of blood-brain barrier and hemorrhagic transformation induced by tissue plasminogen activator in cerebral ischemia. Neurobiol Dis 2011; 44:277-83. [PMID: 21816222 DOI: 10.1016/j.nbd.2011.07.004] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2011] [Revised: 06/24/2011] [Accepted: 07/06/2011] [Indexed: 10/18/2022] Open
Abstract
Tissue plasminogen activator (tPA) remains the only approved thrombolytic agent for the early treatment of ischemic stroke. However, treatment with tPA may lead to disruption of the blood-brain barrier and hemorrhagic transformation. 17β-estradiol (E2) has demonstrated efficacy in reduction of infarct volume in ischemic stroke models. The effects of acute administration of E2 on permeability of the blood-brain barrier and its ability to prevent hemorrhagic transformation in ischemic rats treated with tPA have not previously been studied. Here, we show that neurological deficits, brain water content, and Evan's blue extravasation were increased in ovariectomized female Wistar rats treated with tPA and attenuated in rats receiving E2+tPA. We also show that intracerebral hemoglobin and matrix metalloproteinase-9 activity were elevated with tPA treatment, and these increases were reduced by E2 treatment. Taken together, these data demonstrate that acute administration of E2 is capable of ameliorating some of the adverse effects of tPA administration, including the increase of matrix metalloproteinase-9 activity, blood-brain barrier permeability, and hemorrhagic transformation. These findings suggest a potential role for estrogen in thrombolytic treatment for ischemic stroke.
Collapse
Affiliation(s)
- Mingchang Li
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | | | | | | | | |
Collapse
|
8
|
Abstract
Evidence exists for the potential protective effects of circulating ovarian hormones in stroke, and oestrogen reduces brain damage in animal ischaemia models. However, a recent clinical trial indicated that HRT (hormone-replacement therapy) increased the incidence of stroke in post-menopausal women, and detrimental effects of oestrogen on stroke outcome have been identified in a meta-analysis of HRT trials and in pre-clinical research studies. Therefore oestrogen is not an agent that can be promoted as a potential stroke therapy. Many published reviews have reported the neuroprotective effects of oestrogen in stroke, but have failed to include information on the detrimental effects. This issue is addressed in the present review, along with potential mechanisms of action, and the translational capacity of pre-clinical research.
Collapse
|
9
|
Pereno G, Beltramino C. Differential role of gonadal hormones on kainic acid–induced neurodegeneration in medial amygdaloid nucleus of female and male rats. Neuroscience 2009; 163:952-63. [DOI: 10.1016/j.neuroscience.2009.06.059] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2009] [Revised: 06/24/2009] [Accepted: 06/27/2009] [Indexed: 11/30/2022]
|
10
|
Bramlett HM, Furones-Alonso O, Lotocki G, Rodriguez-Paez A, Sanchez-Molano J, Keane RW. Sex differences in XIAP cleavage after traumatic brain injury in the rat. Neurosci Lett 2009; 461:49-53. [PMID: 19500649 PMCID: PMC2716731 DOI: 10.1016/j.neulet.2009.05.071] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2009] [Revised: 05/19/2009] [Accepted: 05/27/2009] [Indexed: 11/27/2022]
Abstract
Sex influences histological and behavioral outcomes following traumatic brain injury (TBI), but the underlying sex-dependent pathomechanisms regulating outcome measures remain poorly defined. Here, we investigated the TBI-induced regulation of the X-linked inhibitor of apoptosis protein (XIAP) that, in addition to suppressing cell death by inhibition of caspases, is involved in signaling cascades, including immune regulation and cell migration. Since estrogen has been shown to have anti-apoptotic properties, we specifically examined sex differences and the influence of estrogen on XIAP processing after TBI. Sprague-Dawley male (TBI-M), female (TBI-F), ovariectomized female (TBI-OVX) and ovariectomized females supplemented with estrogen (TBI-OVX+EST) were subjected to moderate (1.7-2.2atm) fluid percussion (FP) injury. Animals were sacrificed 24h after FP injury; cortical tissue (ipsilateral and contralateral) was dissected and analyzed for XIAP processing by immunoblot analysis (n=6-7/group) or confocal microscopy (n=2-3/group). Significant differences in XIAP cleavage products in the ipsilateral cortex were found between groups (p<0.03). Post hoc analysis showed an increase in XIAP processing in both TBI-F and TBI-OVX+EST compared to TBI-M and TBI-OVX (p<0.05), indicating that more XIAP is cleaved following injury in intact females and TBI-OVX+EST than in TBI-M and TBI-OVX groups. Co-localization of XIAP within neurons also demonstrated sex-dependent changes. Based on these data, it appears that the processing of XIAP after injury is different between males and females and may be influenced by exogenous estrogen treatment.
Collapse
Affiliation(s)
- Helen M Bramlett
- Department of Neurological Surgery, Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33101, USA.
| | | | | | | | | | | |
Collapse
|
11
|
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
|
12
|
Farr TD, Carswell HVO, McCann DJ, Sato M, Bryant HU, Dodge JA, Macrae IM. The selective oestrogen receptor modulator, LY362321, is not neuroprotective in a rat model of transient focal ischaemia. J Neuroendocrinol 2008; 20:366-74. [PMID: 18208545 DOI: 10.1111/j.1365-2826.2008.01648.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Selective oestrogen receptor modulators (SERMs) may offer improved alternatives to oestrogen as neuroprotectants in experimental stroke. The present study investigated the role of a novel SERM, LY362321, in a rat model of transient middle cerebral artery occlusion (MCAO). Female Sprague-Dawley rats were ovariectomised and began receiving daily s.c. injections of either 1 mg/kg (n = 13), 10 mg/kg (n = 14) of LY362321, or vehicle (n = 13). The left MCA was temporarily occluded (90 min), with cortical blood flow monitoring, at 12 days post ovariectomy. Sensorimotor function was assessed using a neurological score prior to the MCAO and daily for 3 days following the MCAO. Tissue was processed for infarct volume assessment using 2,3,5-triphenyltetra-zolium chloride staining. The results indicated that there were no significant differences amongst groups in cortical blood flow during the MCAO. Furthermore, there was no significant difference in infarct size amongst vehicle, 1, and 10 mg/kg treated animals: 22.9 +/- 5.0, 16.7 +/- 4.2, and 21.1 +/- 4.1, respectively, one-way anova [F(2,32) = 0.542, P = 0.587]. The MCAO induced a significant decline in neurological score in the vehicle group (from 14 to 7 at 24 h post-MCAO) but this was not significantly affected by LY362321 at either dose. In conclusion, pretreatment with a low or high dose of the novel SERM LY362321 did not significantly influence cerebral blood flow, infarct volume, or sensorimotor function in rats exposed to transient MCAO.
Collapse
Affiliation(s)
- T D Farr
- Wellcome Surgical Institute and 7T MRI Facility, Division of Clinical Neuroscience, University of Glasgow, Glasgow, UK.
| | | | | | | | | | | | | |
Collapse
|
13
|
Obrenovitch TP. Molecular physiology of preconditioning-induced brain tolerance to ischemia. Physiol Rev 2008; 88:211-47. [PMID: 18195087 DOI: 10.1152/physrev.00039.2006] [Citation(s) in RCA: 176] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Ischemic tolerance describes the adaptive biological response of cells and organs that is initiated by preconditioning (i.e., exposure to stressor of mild severity) and the associated period during which their resistance to ischemia is markedly increased. This topic is attracting much attention because preconditioning-induced ischemic tolerance is an effective experimental probe to understand how the brain protects itself. This review is focused on the molecular and related functional changes that are associated with, and may contribute to, brain ischemic tolerance. When the tolerant brain is subjected to ischemia, the resulting insult severity (i.e., residual blood flow, disruption of cellular transmembrane gradients) appears to be the same as in the naive brain, but the ensuing lesion is substantially reduced. This suggests that the adaptive changes in the tolerant brain may be primarily directed against postischemic and delayed processes that contribute to ischemic damage, but adaptive changes that are beneficial during the subsequent test insult cannot be ruled out. It has become clear that multiple effectors contribute to ischemic tolerance, including: 1) activation of fundamental cellular defense mechanisms such as antioxidant systems, heat shock proteins, and cell death/survival determinants; 2) responses at tissue level, especially reduced inflammatory responsiveness; and 3) a shift of the neuronal excitatory/inhibitory balance toward inhibition. Accordingly, an improved knowledge of preconditioning/ischemic tolerance should help us to identify neuroprotective strategies that are similar in nature to combination therapy, hence potentially capable of suppressing the multiple, parallel pathophysiological events that cause ischemic brain damage.
Collapse
Affiliation(s)
- Tihomir Paul Obrenovitch
- Division of Pharmacology, School of Life Sciences, University of Bradford, Bradford, United Kingdom.
| |
Collapse
|
14
|
McCullough LD, Koerner IP, Hurn PD. Effects of gender and sex steroids on ischemic injury. HANDBOOK OF CLINICAL NEUROLOGY 2008; 92:149-69. [PMID: 18790274 DOI: 10.1016/s0072-9752(08)01908-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
15
|
Lovekamp-Swan T, Glendenning M, Schreihofer DA. A high soy diet reduces programmed cell death and enhances bcl-xL expression in experimental stroke. Neuroscience 2007; 148:644-52. [PMID: 17706879 PMCID: PMC2788970 DOI: 10.1016/j.neuroscience.2007.06.046] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2007] [Revised: 06/13/2007] [Accepted: 07/20/2007] [Indexed: 10/23/2022]
Abstract
Soy phytoestrogens have been proposed as an alternative to estrogen replacement therapy and have demonstrated potential neuroprotective effects in the brain. We have shown that a high soy diet significantly reduces infarct size following permanent middle cerebral artery occlusion (MCAO). Here, we tested the hypothesis that a high soy diet would attenuate programmed cell death after stroke. Adult female Sprague-Dawley rats were ovariectomized and fed either an isoflavone-reduced diet (IFP) or a high soy diet (SP) for 2 weeks before undergoing 90 min of transient middle cerebral artery occlusion (tMCAO) followed by 22.5 h reperfusion. Infarct size, as assessed by triphenyltetrazolium chloride staining, was significantly reduced by a high soy diet (P<0.05). Apoptosis in the ischemic cortex, measured by TUNEL staining, was significantly reduced by the high soy diet. The number of active caspase-3 positive cells and caspase-mediated alpha-spectrin cleavage were also significantly decreased in the ischemic cortex of SP rats. Furthermore, nuclear translocation of apoptosis-inducing factor (AIF) was significantly reduced in the ischemic cortex of SP rats. Soy significantly increased bcl-x(L) mRNA and protein expression in the ischemic cortex compared with IFP rats. Immunohistochemistry revealed increased neuronal expression of bcl-2 and bcl-x(L) in the ischemic cortex of both IFP and SP rats following tMCAO. These results suggest that a high soy diet decreases both caspase-dependent and caspase-independent programmed cell death following tMCAO. Further, a high soy diet enhances expression of the cell survival factor bcl-x(L) following tMCAO, contributing to the neuroprotective effects of soy in the ischemic cortex.
Collapse
MESH Headings
- Active Transport, Cell Nucleus/drug effects
- Active Transport, Cell Nucleus/genetics
- Animals
- Apoptosis/drug effects
- Apoptosis/genetics
- Apoptosis Inducing Factor/drug effects
- Apoptosis Inducing Factor/genetics
- Apoptosis Inducing Factor/metabolism
- Apoptosis Regulatory Proteins/drug effects
- Apoptosis Regulatory Proteins/genetics
- Apoptosis Regulatory Proteins/metabolism
- Brain/drug effects
- Brain/metabolism
- Brain/physiopathology
- Caspases/drug effects
- Caspases/genetics
- Caspases/metabolism
- Cell Survival/drug effects
- Cell Survival/genetics
- Disease Models, Animal
- Estrogens/metabolism
- Female
- Food, Formulated
- Infarction, Middle Cerebral Artery/drug therapy
- Infarction, Middle Cerebral Artery/genetics
- Infarction, Middle Cerebral Artery/metabolism
- Nerve Degeneration/drug therapy
- Nerve Degeneration/genetics
- Nerve Degeneration/metabolism
- Neuroprotective Agents/metabolism
- Neuroprotective Agents/pharmacology
- Neuroprotective Agents/therapeutic use
- Phytoestrogens/metabolism
- Phytoestrogens/pharmacology
- Phytoestrogens/therapeutic use
- Proto-Oncogene Proteins c-bcl-2/drug effects
- Proto-Oncogene Proteins c-bcl-2/genetics
- Proto-Oncogene Proteins c-bcl-2/metabolism
- RNA, Messenger/drug effects
- RNA, Messenger/metabolism
- Rats
- Rats, Sprague-Dawley
- Soy Foods
- Stroke/drug therapy
- Stroke/genetics
- Stroke/metabolism
- bcl-X Protein/drug effects
- bcl-X Protein/genetics
- bcl-X Protein/metabolism
Collapse
Affiliation(s)
- T Lovekamp-Swan
- Department of Physiology, CA3145, Medical College of Georgia, 1120 15th Street, Augusta, GA 30912, USA
| | | | | |
Collapse
|
16
|
Kaur P, Jodhka PK, Underwood WA, Bowles CA, de Fiebre NC, de Fiebre CM, Singh M. Progesterone increases brain-derived neuroptrophic factor expression and protects against glutamate toxicity in a mitogen-activated protein kinase- and phosphoinositide-3 kinase-dependent manner in cerebral cortical explants. J Neurosci Res 2007; 85:2441-9. [PMID: 17549730 PMCID: PMC2693123 DOI: 10.1002/jnr.21370] [Citation(s) in RCA: 138] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The higher prevalence and risk for Alzheimer's disease in women relative to men has been partially attributed to the precipitous decline in gonadal hormone levels that occurs in women following the menopause. Although considerable attention has been focused on the consequence of estrogen loss, and thus estrogen's neuroprotective potential, it is important to recognize that the menopause results in a precipitous decline in progesterone levels as well. In fact, progesterone is neuroprotective, although the precise mechanisms involved remain unclear. Based on our previous observation that progesterone elicits the phosphorylation of ERK and Akt, key effectors of the neuroprotective mitogen-activated protein kinase (MAPK) and phosphoinositide-3 kinase (PI3-K) pathways, respectively, we determined whether activation of either of these pathways was necessary for progesterone-induced protection. With organotypic explants (slice culture) of the cerebral cortex, we found that progesterone protected against glutamate-induced toxicity. Furthermore, these protective effects were inhibited by either the MEK1/2 inhibitor UO126 or the PI3-K inhibitor LY294002, supporting the requirement for both the MAPK and PI3-K pathways in progesterone-induced protection. In addition, at a concentration and duration of treatment consistent with our neuroprotection data, progesterone also increased the expression of brain-derived neurotrophic factor (BDNF), at the level of both protein and mRNA. This induction of BDNF may be relevant to the protective effects of progesterone, in that inhibition of Trk signaling, with K252a, inhibited the protective effects of progesterone. Collectively, these data suggest that progesterone is protective via multiple and potentially related mechanisms. (c) 2007 Wiley-Liss, Inc.
Collapse
Affiliation(s)
- Paramjit Kaur
- Department of Pharmacology & Neuroscience and the Institute for Aging & Alzheimer’s Disease Research, The Center FOR HER (Focused on Resources for her Health Education and Research), University of North Texas Health Science Center, Fort Worth, Texas
| | - Parmeet K. Jodhka
- Department of Pharmacology & Neuroscience and the Institute for Aging & Alzheimer’s Disease Research, The Center FOR HER (Focused on Resources for her Health Education and Research), University of North Texas Health Science Center, Fort Worth, Texas
| | - Wendy A. Underwood
- Department of Pharmacology & Neuroscience and the Institute for Aging & Alzheimer’s Disease Research, The Center FOR HER (Focused on Resources for her Health Education and Research), University of North Texas Health Science Center, Fort Worth, Texas
| | - Courtney A. Bowles
- Department of Pharmacology & Neuroscience and the Institute for Aging & Alzheimer’s Disease Research, The Center FOR HER (Focused on Resources for her Health Education and Research), University of North Texas Health Science Center, Fort Worth, Texas
| | - NancyEllen C. de Fiebre
- Department of Pharmacology & Neuroscience and the Institute for Aging & Alzheimer’s Disease Research, The Center FOR HER (Focused on Resources for her Health Education and Research), University of North Texas Health Science Center, Fort Worth, Texas
| | - Christopher M. de Fiebre
- Department of Pharmacology & Neuroscience and the Institute for Aging & Alzheimer’s Disease Research, The Center FOR HER (Focused on Resources for her Health Education and Research), University of North Texas Health Science Center, Fort Worth, Texas
| | - Meharvan Singh
- Department of Pharmacology & Neuroscience and the Institute for Aging & Alzheimer’s Disease Research, The Center FOR HER (Focused on Resources for her Health Education and Research), University of North Texas Health Science Center, Fort Worth, Texas
| |
Collapse
|
17
|
Gibson CL, Gray LJ, Murphy SP, Bath PMW. Estrogens and experimental ischemic stroke: a systematic review. J Cereb Blood Flow Metab 2006; 26:1103-13. [PMID: 16437060 DOI: 10.1038/sj.jcbfm.9600270] [Citation(s) in RCA: 115] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Estrogens are believed to provide females with endogenous protection against cerebrovascular events although clinical trials studying long-term hormone replacement have yielded disappointing results. In contrast, estrogens might be neuroprotective after experimental ischemia. We performed a systematic review of controlled experimental studies that administered estrogens before, or after, cerebral ischemia and measured lesion volume. Relevant studies were found from searching PubMed, Embase and Web of Science. From 161 identified publications, 27 studies using 1,304 experimental subjects were analyzed using the Cochrane Review Manager software. Estrogens reduced lesion volume in a dose-dependent manner, after either transient (P<0.001) or permanent (P<0.001) ischemia and whether administered before or up to 4 h after ischemia onset; no studies assessed efficacy for later time periods. The effect size for estrogens decreased with increasing quality scores for studies of transient ischemia. Estrogens reduced lesion volume when administered to ovariectomized females and young adult males, but had no effect in intact females. Limited data were present for aged animals and the full dose-response relationship was not available in all experimental groups. On the basis of these data, estrogens are a candidate treatment for ischemic stroke, although further preclinical studies are also warranted.
Collapse
Affiliation(s)
- Claire L Gibson
- Institute of Cell Signalling, Queen's Medical Centre, University of Nottingham, Nottingham, UK.
| | | | | | | |
Collapse
|
18
|
Farr TD, Carswell HVO, Gallagher L, Condon B, Fagan AJ, Mullin J, Macrae IM. 17β-Estradiol treatment following permanent focal ischemia does not influence recovery of sensorimotor function. Neurobiol Dis 2006; 23:552-62. [PMID: 16759876 DOI: 10.1016/j.nbd.2006.04.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2006] [Revised: 04/07/2006] [Accepted: 04/24/2006] [Indexed: 11/23/2022] Open
Abstract
The development of therapy to aid poststroke recovery is essential. The female hormone 17beta-estradiol has been shown to promote synaptogenesis; the purpose of this study was to attempt to harness these mechanisms to promote repair and recovery in the peri-infarct zone. Rats were ovariectomized, tested for sensorimotor function, and the middle cerebral artery permanently occluded (MCAO). Infarct volumes were calculated using MRI, and damage was equivalent in all animals prior to implantation of either 17beta-estradiol or placebo pellets. Animals were tested for functional recovery for 28 days and tissue processed for synaptic marker syntaxin immunohistochemistry. The stroke induced a significant behavioral deficit, which persisted out to 28 days, and was not significantly different between 17beta-estradiol and placebo treatment groups. There was no difference in syntaxin immunostaining between groups in either the peri-infarct cortex or in the dendritic CA1 reference region. In conclusion, 17beta-estradiol treatment, delivered poststroke, did not influence recovery of function or synaptogenesis.
Collapse
Affiliation(s)
- Tracy D Farr
- 7TMRI Facility and Wellcome Surgical Institute, Division of Clinical Neuroscience, University of Glasgow, Garscube Estate, Bearsden Road, Glasgow, Scotland G61 1QH, UK.
| | | | | | | | | | | | | |
Collapse
|
19
|
Johnson AB, Bake S, Lewis DK, Sohrabji F. Temporal expression of IL-1beta protein and mRNA in the brain after systemic LPS injection is affected by age and estrogen. J Neuroimmunol 2006; 174:82-91. [PMID: 16530273 DOI: 10.1016/j.jneuroim.2006.01.019] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2005] [Revised: 01/19/2006] [Accepted: 01/27/2006] [Indexed: 11/17/2022]
Abstract
Estrogen has been shown to suppress neural inflammation in vivo in response to intracerebral LPS injections or by intraparenchymal injections of NMDA. Using the latter approach, we have shown that estrogen suppresses inflammatory cytokine expression in lesioned ovariectomized young adult females but not reproductive senescent animals. However, in cultured microglia derived from either young or senescent animals, estrogen fails to suppress LPS-induced cytokine expression. These data suggest that estrogen's effects on the neural inflammatory response may result from its actions on blood-borne immune cells or its actions at the blood brain barrier or both. This hypothesis was directly tested here using a systemic injury model and comparing the neural inflammatory response in the olfactory bulb, which is protected by the blood brain barrier, and in the pituitary gland, which is incompletely protected by the blood brain barrier. Young and senescent Sprague-Dawley female rats were ovariectomized and replaced with either an estrogen or placebo pellet. Three weeks later, animals received a single i.p. injection of LPS (or vehicle) and were terminated 0.5, 2 or 3h later. Systemic injections of LPS increased IL-1beta expression in the liver in a time-dependent manner in young and senescent females. In young adults, LPS increased cytokine expression in both the bulb and the pituitary gland. However, estrogen treatment attenuated IL-1beta expression in the olfactory bulb but not in the pituitary gland. In senescent animals, estrogen completely suppressed IL-1beta expression in the bulb and the pituitary gland, while placebo-replaced animals responded normally. This age-related difference in cytokine induction by LPS was also seen in mRNA regulation, such that LPS induced IL-1beta mRNA in the olfactory bulb of young adults but not in the senescent female. Age and hormone effects on pituitary cytokines were also mirrored in plasma corticosterone (CORT) levels, such that estrogen treatment to senescent females attenuated LPS-induced CORT. These data suggest that the central inflammatory response to a systemic insult can be modulated by estrogen although the mechanism underlying the initiation of this response varies with reproductive age.
Collapse
Affiliation(s)
- Adam B Johnson
- Department of Anatomy and Medical Neurobiology, TAMUS Health Science Center, 228 Reynolds Medical Building, College Station, TX 77843-1114, USA
| | | | | | | |
Collapse
|
20
|
Liu R, Wen Y, Perez E, Wang X, Day AL, Simpkins JW, Yang SH. 17β-Estradiol attenuates blood–brain barrier disruption induced by cerebral ischemia–reperfusion injury in female rats. Brain Res 2005; 1060:55-61. [PMID: 16212944 DOI: 10.1016/j.brainres.2005.08.048] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2005] [Revised: 08/16/2005] [Accepted: 08/17/2005] [Indexed: 11/26/2022]
Abstract
Disruption of blood-brain barrier (BBB), mediated through matrix metalloproteinases (MMPs), is a critical event during cerebral ischemia. While neuroprotective effects of estrogens have been well established in ischemic stroke models, the effects of estrogens on BBB integrity remain to be elucidated. In the present study, we determined effects of 17beta-estradiol (E2) on BBB disruption induced by transient focal cerebral ischemia and its effects on MMP2 and MMP9 activation. Transient cerebral ischemia was induced by middle cerebral artery (MCA) occlusion for 1 h followed by reperfusion in ovariectomized rats. E2 (100 microg/kg) or vehicle was administered 2 h before MCA occlusion. BBB integrity was determined by fluorescent detection of extravasated Evans blue. In separate experiments, effect of E2 on MMP2 and MMP9 expression and activation was determined by immunoblot and MMPs activity assay. E2 treatment prevented more than 50% and 30% of BBB disruption in the ischemic cortex and subcortex at 4 h after reperfusion, respectively. MMP2 and MMP9 expression was elevated at 2 h and peaked at 4 h after reperfusion in the ischemic cortex, which was markedly reduced by E2 treatment. E2 treatment also attenuated the increase of MMPs activity induced by ischemia-reperfusion injury. In conclusion, estrogens could attenuate BBB disruption induced by transient cerebral ischemia, by inhibition of MMP2 and MMP9 activation. Our results suggest an important role of estrogens as multiple targeting protectants against ischemic stroke on cellular as well as vascular components of central nervous system.
Collapse
Affiliation(s)
- Ran Liu
- Department of Pharmacology and Neuroscience, University of North Texas, Health Science Center at Fort Worth, 3500 Camp Bowie Boulevard, Fort Worth, TX 76107, USA
| | | | | | | | | | | | | |
Collapse
|
21
|
Bramlett HM. Sex differences and the effect of hormonal therapy on ischemic brain injury. ACTA ACUST UNITED AC 2005; 12:17-27. [PMID: 15927821 DOI: 10.1016/j.pathophys.2005.02.009] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2004] [Revised: 02/01/2005] [Accepted: 02/01/2005] [Indexed: 11/24/2022]
Abstract
Epidemiological data emphasize the importance of sex differences in the mortality and morbidity of stroke and cardiovascular disease. The importance of hormonal influences on stroke outcome has pointed out the importance of gender, age, and presence of neural hormones. This clinical data has been substantiated by various experimental studies using clinically relevant models of cerebral ischemia and stroke. Published findings emphasize that male and female animals respond differently to periods of cerebral ischemia and that various combinations of hormonal treatments can provide protection, both histopathological and behavioral. Mechanisms underlying the hormonal effects on ischemic outcome are multifactorial. These include effects on vascular integrity and cerebral blood flow, excitotoxicity, oxidation pathways, inflammation, and apoptosis. Although many studies have shown positive results with hormonal treatments, negative findings have also been presented. Explanations for the limitations of hormonal treatment include uncertainties regarding therapeutic window, specific therapeutic dose range, as well as the specific pathophysiological processes being targeted. Additional studies are therefore required to clarify under what conditions hormonal therapy is most protective or not warranted. Experimental studies utilizing a variety of cerebral ischemia and stroke models are reviewed to indicate under what conditions sex differences and hormonal therapy are most important in terms of functional outcome.
Collapse
Affiliation(s)
- Helen M Bramlett
- Department of Neurological Surgery, Miami Project to Cure Paralysis, University of Miami School of Medicine, 1095 NW 14th Terrace, Miami, FL 33136, USA
| |
Collapse
|
22
|
Theodorsson A, Holm L, Theodorsson E. Modern anesthesia and peroperative monitoring methods reduce per- and postoperative mortality during transient occlusion of the middle cerebral artery in rats. ACTA ACUST UNITED AC 2005; 14:181-90. [PMID: 15795172 DOI: 10.1016/j.brainresprot.2005.01.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2004] [Revised: 01/25/2005] [Accepted: 01/27/2005] [Indexed: 11/15/2022]
Abstract
Mortality and morbidity during and after occlusion of the middle cerebral artery in rats are important confounding factors which may be minimized by improved anesthesia and peroperative monitoring techniques. We describe state of the art techniques for inducing anesthesia, endotracheal intubation, ventilation and monitoring peroperatively in this context. Introducing the subtemporal approach of Tamura et al. in our laboratory 5 years ago, we experienced 25% peroperative and 24 h postoperative rat mortality when performing temporary clipping of the middle cerebral artery. This prompted us to abandon intraperitoneal anesthesia by chloral hydrate and ventilation by tracheotomy in favor of endotracheal intubation and isoflurane anesthesia (1% isoflurane in 30%:70% O(2)/N(2)O). These anesthetic techniques in combination with improved surgical skills have reduced our initial 25% peroperative- and 24 h postoperative mortality to 2.7% (1.8% peroperatively and 0.9% 24 h postoperatively). Furthermore, the following 14 days postoperative mortality rate was 1.8%. A total number of 203 rats have been operated with this method in different studies where a focal reperfusion stroke model combined with extended periods of observations were the cornerstone.
Collapse
MESH Headings
- Anesthesia, Inhalation/methods
- Anesthetics, Inhalation/pharmacology
- Animals
- Disease Models, Animal
- Female
- Infarction, Middle Cerebral Artery/mortality
- Infarction, Middle Cerebral Artery/pathology
- Infarction, Middle Cerebral Artery/physiopathology
- Intubation, Intratracheal/instrumentation
- Intubation, Intratracheal/methods
- Isoflurane/pharmacology
- Monitoring, Intraoperative/methods
- Postoperative Complications/mortality
- Postoperative Complications/prevention & control
- Rats
- Rats, Sprague-Dawley
- Surgical Instruments
Collapse
Affiliation(s)
- Annette Theodorsson
- Department of Biomedicine and Surgery, Division of Clinical Chemistry, University Hospital, SE-581 85 Linkoping, Sweden.
| | | | | |
Collapse
|
23
|
Dykens JA, Carroll AK, Wiley S, Covey DF, Cai ZY, Zhao L, Wen R. Photoreceptor preservation in the S334ter model of retinitis pigmentosa by a novel estradiol analog. Biochem Pharmacol 2005; 68:1971-84. [PMID: 15476668 DOI: 10.1016/j.bcp.2004.06.042] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2004] [Accepted: 06/15/2004] [Indexed: 11/29/2022]
Abstract
The cytoprotective activity of MITO-4565, a novel, non-hormonal, estradiol derivative, was evaluated in the S334ter transgenic model of retinitis pigmentosa (RP). Progressive blindness in RP is due to apoptotic death of the photoreceptors, a process mimicked by the animal models [Portera-Cailliau C, Sung C-H, Nathans J, Adler R. Apoptotic photoreceptor cell death in mouse models of retinitis pigmentosa. Proc Natl Acad Sci USA 1994;91:974-8]. On postnatal day 9, 10 transgenic S334ter rats received a single intraocular injection of MITO-4565 in the left eye, and vehicle in the right eye. By postnatal day 20, the thickness of the outer nuclear layer (ONL) in the superior retina of the untreated eyes was 5.76 +/- 1.12 microm (N = 10), versus 10.72 +/- 1.52 microm (N = 10) for eyes treated with MITO-4565 (P < 0.0001, ANOVA F = 1671). Comparable cytoprotection was also observed for the inferior retina. Cytoprotection by MITO-4565 was also observed in primary cultures of rat retinal ganglion cells against NMDA excitotoxicity. Data from studies of hexose monophosphate shunt flux, mitochondrial stability, and in vitro lipid peroxidation, are in accord with previous reports [Green PS, Gridley KE, Simpkins JW. Nuclear estrogen receptor independent neuroprotection by estratrienes: a novel interaction with glutathione. Neuroscience 1997;84:7-10]; a likely mechanism of action entails moderation of membrane lipid peroxidation in a redox couple with glutathione. Such preservation of membrane integrity is particularly crucial to mitochondria, where collapse of membrane potential precipitates cell death, and where GSH is maintained at mM concentrations. Indeed, exposure to MITO-4565, but not a methoxy substituted negative control, allowed mitochondria to retain membrane potential (DeltaPsi(m)) under conditions of Ca(2+) overload that would normally induce complete mitochondrial failure. Mitochondrial interventions offer a novel therapeutic approach for RP, and other degenerative diseases of the retina.
Collapse
Affiliation(s)
- James A Dykens
- MitoKor Inc., 11494 Sorrento Valley Road, San Diego, CA 92121, USA.
| | | | | | | | | | | | | |
Collapse
|