1
|
Ally A, Powell I, Ally MM, Chaitoff K, Nauli SM. Role of neuronal nitric oxide synthase on cardiovascular functions in physiological and pathophysiological states. Nitric Oxide 2020; 102:52-73. [PMID: 32590118 DOI: 10.1016/j.niox.2020.06.004] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 03/15/2020] [Accepted: 06/15/2020] [Indexed: 12/16/2022]
Abstract
This review describes and summarizes the role of neuronal nitric oxide synthase (nNOS) on the central nervous system, particularly on brain regions such as the ventrolateral medulla (VLM) and the periaqueductal gray matter (PAG), and on blood vessels and the heart that are involved in the regulation and control of the cardiovascular system (CVS). Furthermore, we shall also review the functional aspects of nNOS during several physiological, pathophysiological, and clinical conditions such as exercise, pain, cerebral vascular accidents or stroke and hypertension. For example, during stroke, a cascade of molecular, neurochemical, and cellular changes occur that affect the nervous system as elicited by generation of free radicals and nitric oxide (NO) from vulnerable neurons, peroxide formation, superoxides, apoptosis, and the differential activation of three isoforms of nitric oxide synthases (NOSs), and can exert profound effects on the CVS. Neuronal NOS is one of the three isoforms of NOSs, the others being endothelial (eNOS) and inducible (iNOS) enzymes. Neuronal NOS is a critical homeostatic component of the CVS and plays an important role in regulation of different systems and disease process including nociception. The functional and physiological roles of NO and nNOS are described at the beginning of this review. We also elaborate the structure, gene, domain, and regulation of the nNOS protein. Both inhibitory and excitatory role of nNOS on the sympathetic autonomic nervous system (SANS) and parasympathetic autonomic nervous system (PANS) as mediated via different neurotransmitters/signal transduction processes will be explored, particularly its effects on the CVS. Because the VLM plays a crucial function in cardiovascular homeostatic mechanisms, the neuroanatomy and cardiovascular regulation of the VLM will be discussed in conjunction with the actions of nNOS. Thereafter, we shall discuss the up-to-date developments that are related to the interaction between nNOS and cardiovascular diseases such as hypertension and stroke. Finally, we shall focus on the role of nNOS, particularly within the PAG in cardiovascular regulation and neurotransmission during different types of pain stimulus. Overall, this review focuses on our current understanding of the nNOS protein, and provides further insights on how nNOS modulates, regulates, and controls cardiovascular function during both physiological activity such as exercise, and pathophysiological conditions such as stroke and hypertension.
Collapse
Affiliation(s)
- Ahmmed Ally
- Arkansas College of Osteopathic Medicine, Fort Smith, AR, USA.
| | - Isabella Powell
- All American Institute of Medical Sciences, Black River, Jamaica
| | | | - Kevin Chaitoff
- Interventional Rehabilitation of South Florida, West Palm Beach, FL, USA
| | - Surya M Nauli
- Chapman University and University of California, Irvine, CA, USA.
| |
Collapse
|
2
|
Estrogen and propofol combination therapy inhibits endoplasmic reticulum stress and remarkably attenuates cerebral ischemia-reperfusion injury and OGD injury in hippocampus. Biomed Pharmacother 2018; 108:1596-1606. [PMID: 30372862 DOI: 10.1016/j.biopha.2018.09.167] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 09/27/2018] [Accepted: 09/28/2018] [Indexed: 01/06/2023] Open
Abstract
AIM Endoplasmic reticulum stress (ERS) is vital in inducing apoptosis via caspase-12 and C/EBP homologous protein (CHOP) apoptotic pathway in the hippocampus after ischemia-reperfusion injury. The study aimed to estimate the efficacy of estrogen and propofol combination therapy against ERS-induced apoptosis after cerebral ischemia-reperfusion injury and oxygen-glucose deprivation (OGD) injury in the hippocampus in vivo and in vitro. METHODS Rat model of cerebral ischemia-reperfusion injury was generated by middle cerebral artery occlusion (MCAO) strategy with ischemic intervention for 90 min and reperfusion for 24 h. Propofol processing ischemia-reperfusion group (Propofol group) infused 50 mg/kg/h of propofol via the femoral vein at the onset of reperfusion for 30 min. Estrogen processing ischemia-reperfusion group (estrogen group) received 0.0125 mg/kg of estrogen via tail vein at 30 min prior to MCAO. Combination therapy for ischemia-reperfusion group (combination group) received simultaneous processing with propofol and estrogen. In vitro, brain slices were randomly exposed to dimethylsulfoxide (DSMO), 10 μm of propofol, 10 nm of estrogen, or propofol and estrogen. Changes in the orthodromic population spike (OPS) at the end of reoxygenation were recorded. Neurological deficit examination, Nissl staining, and 2,3,5-triphenyltetrazolium chloride (TTC) staining were employed to evaluate the level of cerebral ischemia-reperfusion injury. The expression of caspase-3, caspase-12, glucose-regulated protein 78 (GRP78), and CHOP were investigated by Western blot and immunofluorescence staining assays. Neural apoptotic rate in hippocampus was detected by the flow cytometry trial. RESULTS Neurological deficit score, infarct volume, the expression of caspase-3 (P < 0.05), caspase-12, GRP78, CHOP, and neural apoptotic rate of I/R group increased markedly (P < 0.01). When obtaining drug treatment, neurological deficit score (P < 0.05), infarct volume, the expression levels of caspase-12 and GRP78, and neural apoptotic rate of the propofol group decreased significantly (P < 0.01). Furthermore, neurological deficit score, infarct volume, expression levels of caspase-3, caspase-12, GRP78, and CHOP (P < 0.05), and neural apoptotic rate decreased in the estrogen group (P < 0.01) and especially in the combination group (P < 0.01). Compared with the propofol group, the neurological deficit score (P < 0.05), infarct volume, caspase-3, caspase-12, GRP78, CHOP, and neural apoptotic rate of the combination group decreased (P < 0.01). Compared with the estrogen group, the infarct volume, caspase-3 (P < 0.05), GRP78, CHOP, and neural apoptotic rate (P < 0.05) of the combination group decreased (P < 0.01). Compared with the propofol group, the infarct volume, caspase-3, caspase-12 (P < 0.05), and GRP78 (P < 0.05) of the estrogen group decreased (P < 0.01). Propofol and estrogen treatment can delay the abolishing time of OPS and increase the recovery rate and amplitude of OPS, compared with OGD group (P < 0.01), especially in the combination therapy (P < 0.01). CONCLUSION The neuroprotection of propofol and estrogen combination therapy inhibited excessive ERS-induced apoptosis against cerebral ischemia-reperfusion injury and OGD injury in the hippocampus of rats. Furthermore, the outcomes demonstrated that combination therapy yielded synergistic effects.
Collapse
|
3
|
Dominguez R, Zitting M, Liu Q, Patel A, Babadjouni R, Hodis DM, Chow RH, Mack WJ. Estradiol Protects White Matter of Male C57BL6J Mice against Experimental Chronic Cerebral Hypoperfusion. J Stroke Cerebrovasc Dis 2018; 27:1743-1751. [PMID: 29602614 PMCID: PMC5972054 DOI: 10.1016/j.jstrokecerebrovasdis.2018.01.030] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 01/03/2018] [Accepted: 01/25/2018] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND AND PURPOSE Estradiol is a sex steroid hormone known to protect the brain against damage related to transient and global cerebral ischemia. In the present study, we leverage an experimental murine model of bilateral carotid artery stenosis (BCAS) to examine the putative effects of estradiol therapy on chronic cerebral hypoperfusion. We hypothesize that long-term estradiol therapy protects against white matter injury and declarative memory deficits associated with chronic cerebral hypoperfusion. METHODS Adult male C57BL/6J mice underwent either surgical BCAS or sham procedures. Two days after surgery, the mice were given oral estradiol (Sham+E, BCAS+E) or placebo (Sham+P, BCAS+P) treatments daily for 31-34 days. All mice underwent Novel Object Recognition (NOR) testing 31-34 days after the start of oral treatments. Following sacrifice, blood was collected and brains fixed, sliced, and prepared for histological examination of white matter injury and extracellular signal-regulated kinase (ERK) expression. RESULTS Animals receiving long-term oral estradiol therapy (BCAS-E2 and Sham-E2) had higher plasma estradiol levels than those receiving placebo treatment (BCAS-P and Sham-P). BCAS-E2 mice demonstrated less white matter injury (Klüver-Barrera staining) and performed better on the NOR task compared to BCAS-P mice. ERK expression in the brain was increased in the BCAS compared to sham cohorts. Among the BCAS mice, the BCAS-E2 cohort had a greater number of ERK + cells. CONCLUSION This study demonstrates a potentially protective role for oral estradiol therapy in the setting of white matter injury and declarative memory deficits secondary to murine chronic cerebral hypoperfusion.
Collapse
Affiliation(s)
- Reymundo Dominguez
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Madison Zitting
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Qinghai Liu
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Arati Patel
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Robin Babadjouni
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Drew M Hodis
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Robert H Chow
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - William J Mack
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, California.
| |
Collapse
|
4
|
Stoop W, De Geyter D, Verachtert S, Brouwers S, Verdood P, De Keyser J, Kooijman R. Post-stroke treatment with 17β-estradiol exerts neuroprotective effects in both normotensive and hypertensive rats. Neuroscience 2017; 348:335-345. [DOI: 10.1016/j.neuroscience.2017.02.040] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Revised: 02/16/2017] [Accepted: 02/17/2017] [Indexed: 12/01/2022]
|
5
|
Lamprecht MR, Morrison B. GPR30 activation is neither necessary nor sufficient for acute neuroprotection by 17β-estradiol after an ischemic injury in organotypic hippocampal slice cultures. Brain Res 2014; 1563:131-7. [DOI: 10.1016/j.brainres.2014.03.037] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Revised: 03/04/2014] [Accepted: 03/25/2014] [Indexed: 12/11/2022]
|
6
|
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
|
7
|
Pabon M, Tamboli C, Tamboli S, Acosta S, De La Pena I, Sanberg PR, Tajiri N, Kaneko Y, Borlongan CV. ESTROGEN REPLACEMENT THERAPY FOR STROKE. CELL MEDICINE 2014; 6:111-122. [PMID: 24999442 DOI: 10.3727/215517913x672263] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Stroke is the third most common cause of death and severe disability among Western populations. Overall, the incidence of stroke is uniformly higher in men than in women. Stroke is rare in women during the reproductive years, and rapidly increases after menopause, strongly suggesting that estrogen (E2) plays an important role in the prevention of stroke. Ongoing studies are currently evaluating both the benefits and risks associated with E2 replacement therapy and hormone replacement therapy in stroke. Equally important is the role of E2 receptor (ER), as studies indicate that ER populations in several tissue sites may significantly change during stress and aging. Such changes may affect the patient's susceptibility to neurological disorders including stroke, and greatly affect the response to selective E2 receptor modulators (SERMs). Replacement therapies may be inefficient with low ER levels. The goal of this review paper is to discuss an animal model that will allow investigations of the potential therapeutic effects of E2 and its derivatives in stroke. We hypothesize that E2 neuroprotection is, in part, receptor mediated. This hypothesis is a proof of principle approach to demonstrate a role for specific ER subtypes in E2 neuroprotection. To accomplish this, we use a retroviral mediated gene transfer strategy that express subtypes of the ER gene in regions of the rat brain most susceptible to neuronal damage, namely the striatum and cortex. The animal model is exposed to experimental stroke conditions involving middle cerebral artery occlusion (MCAo) method, and eventually the extent of neuronal damage will be evaluated. A reduction in neuronal damage is expected when E2 is administered with specific ER subtypes. From this animal model, an optimal E2 dose and treatment regimen can be determined. The animal model can help identify potential E2-like therapeutics in stroke, and screen for beneficial or toxic additives present in commercial E2 preparations that are currently available. Such studies will be informative in designing drug therapies for stroke.
Collapse
Affiliation(s)
- Mibel Pabon
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, 12901 Bruce B. Downs Blvd., Tampa, FL USA
| | - Cyrus Tamboli
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, 12901 Bruce B. Downs Blvd., Tampa, FL USA
| | - Sarosh Tamboli
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, 12901 Bruce B. Downs Blvd., Tampa, FL USA
| | - Sandra Acosta
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, 12901 Bruce B. Downs Blvd., Tampa, FL USA
| | - Ike De La Pena
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, 12901 Bruce B. Downs Blvd., Tampa, FL USA
| | - Paul R Sanberg
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, 12901 Bruce B. Downs Blvd., Tampa, FL USA
| | - Naoki Tajiri
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, 12901 Bruce B. Downs Blvd., Tampa, FL USA
| | - Yuji Kaneko
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, 12901 Bruce B. Downs Blvd., Tampa, FL USA
| | - Cesar V Borlongan
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, 12901 Bruce B. Downs Blvd., Tampa, FL USA
| |
Collapse
|
8
|
Ma YL, Qin P, Li Y, Shen L, Wang SQ, Dong HL, Hou WG, Xiong LZ. The effects of different doses of estradiol (E2) on cerebral ischemia in an in vitro model of oxygen and glucose deprivation and reperfusion and in a rat model of middle carotid artery occlusion. BMC Neurosci 2013; 14:118. [PMID: 24106772 PMCID: PMC3851874 DOI: 10.1186/1471-2202-14-118] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Accepted: 09/25/2013] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND Because neuroprotective effects of estrogen remain controversial, we aimed to investigate the effect of different doses of estradiol (E2) on cerebral ischemia using both in vivo and in vitro experiments. RESULTS PC12 cells were cultured at physiological (10 nM and 20 nM) or pharmacological (10 μM and 20 μM) dosages of E2 for 24 hours (h). The results of 5-bromodeoxyuridine (Brdu) incorporation and flow cytometric analysis showed that physiological doses of E2 enhanced cell proliferation and pharmacological doses of E2 inhibited cell proliferation. After the cells were exposed to oxygen and glucose deprivation (OGD) for 4 h and reperfusion for 20 h, the results of 3-(4, 5-dimethylthiazol-2-yl) 2, 5-diphenyl tetrazolium bromide (MTT) assay, lactate dehydrogenase (LDH) release assay, flow cytometric analysis and Western blot analysis showed that physiological doses of E2 enhanced cell viability, reduced cell apoptosis and decreased the expression of pro-apoptotic protein caspase-3. In contrast, pharmacological doses of E2 decreased cell viability and induced cell apoptosis. In vivo, adult ovariectomized (OVX) female rats received continuous subcutaneous injection of different doses of E2 for 4 weeks. Transient cerebral ischemia was induced for 2 h using the middle cerebral artery occlusion (MCAO) technique, followed by 22 h of reperfusion. The results of Garcia test, 2, 3, 5-triphenyltetrazolium chloride (TTC) staining showed that 6 μg/kg and 20 μg/kg E2 replacement induced an increase in neurological deficit scores, a decrease in the infarct volume and a reduction in the expression of caspase-3 when compared to animals in the OVX group without E2 treatment. However, 50 μg/kg E2 replacement treatment decreased neurological deficit scores, increased the infarct volume and the expression of caspase-3 when compared to animals in the control group and 6 up/kg or 20 μg/kg E2 replacement group. CONCLUSION We conclude that physiological levels of E2 exhibit neuroprotective effects on cerebral ischemia; whereas, pharmacological or supraphysiological doses of E2 have damaging effects on neurons after cerebral ischemia.
Collapse
Affiliation(s)
- Yu-Long Ma
- Department of Anesthesiology, Xijing Hospital, The Fourth Military Medical University, Xi’an 710032, P R China
| | - Pei Qin
- Department of Anesthesiology, Xijing Hospital, The Fourth Military Medical University, Xi’an 710032, P R China
| | - Yan Li
- Department of Anesthesiology, Xijing Hospital, The Fourth Military Medical University, Xi’an 710032, P R China
- Department of Biochemistry and Molecular Biology, The State Key Laboratory of Cancer Biology, The Fourth Military Medical University, Xi’an 710032, P R China
| | - Lan Shen
- Department of Biochemistry and Molecular Biology, The State Key Laboratory of Cancer Biology, The Fourth Military Medical University, Xi’an 710032, P R China
| | - Shi-Quan Wang
- Department of Anesthesiology, Xijing Hospital, The Fourth Military Medical University, Xi’an 710032, P R China
| | - Hai-Long Dong
- Department of Anesthesiology, Xijing Hospital, The Fourth Military Medical University, Xi’an 710032, P R China
| | - Wu-Gang Hou
- Department of Anesthesiology, Xijing Hospital, The Fourth Military Medical University, Xi’an 710032, P R China
| | - Li-Ze Xiong
- Department of Anesthesiology, Xijing Hospital, The Fourth Military Medical University, Xi’an 710032, P R China
| |
Collapse
|
9
|
Hypercapnic Chemosensitivity in Patients with Heart Failure: Relation to Shifts in Type-1 Insulin-Like Growth Factor and Sex Hormone-Binding Globulin Levels. NEUROPHYSIOLOGY+ 2013. [DOI: 10.1007/s11062-013-9348-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
10
|
Huang SS, Su HH, Kuo TB, Chen CY, Lan YY, Liu BY, Yang DI, Tsai SC, Yang CC. Suppressing cardiac vagal modulation and changing sleep patterns in rats after chronic ischemic stroke injury. Auton Neurosci 2012; 169:116-23. [DOI: 10.1016/j.autneu.2012.05.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2011] [Revised: 05/28/2012] [Accepted: 05/30/2012] [Indexed: 10/28/2022]
|
11
|
Ström JO, Theodorsson A, Ingberg E, Isaksson IM, Theodorsson E. Ovariectomy and 17β-estradiol replacement in rats and mice: a visual demonstration. J Vis Exp 2012:e4013. [PMID: 22710371 DOI: 10.3791/4013] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Estrogens are a family of female sexual hormones with an exceptionally wide spectrum of effects. When rats and mice are used in estrogen research they are commonly ovariectomized in order to ablate the rapidly cycling hormone production, replacing the 17β-estradiol exogenously. There is, however, lack of consensus regarding how the hormone should be administered to obtain physiological serum concentrations. This is crucial since the 17β-estradiol level/administration method profoundly influences the experimental results. We have in a series of studies characterized the different modes of 17β-estradiol administration, finding that subcutaneous silastic capsules and per-oral nut-cream Nutella are superior to commercially available slow-release pellets (produced by the company Innovative Research of America) and daily injections in terms of producing physiological serum concentrations of 17β-estradiol. Amongst the advantages of the nut-cream method, that previously has been used for buprenorphine administration, is that when used for estrogen administration it resembles peroral hormone replacement therapy and is non-invasive. The subcutaneous silastic capsules are convenient and produce the most stable serum concentrations. This video article contains step-by-step demonstrations of ovariectomy and 17β-estradiol hormone replacement by silastic capsules and peroral Nutella in rats and mice, followed by a discussion of important aspects of the administration procedures.
Collapse
Affiliation(s)
- Jakob O Ström
- Clinical Chemistry and Neurosurgery, Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University.
| | | | | | | | | |
Collapse
|
12
|
Novel Neurovascular Protective Agents: Effects of INV-155, INV-157, INV-159, and INV-161 versus Lipoic Acid and Captopril in a Rat Stroke Model. Cardiol Res Pract 2012; 2012:319230. [PMID: 22263115 PMCID: PMC3259480 DOI: 10.1155/2012/319230] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2011] [Revised: 09/19/2011] [Accepted: 10/01/2011] [Indexed: 02/05/2023] Open
Abstract
Background. Lipoic acid (LA), which has significant antioxidant properties, may also function as a potent neuroprotectant. The synthetic compounds INV-155, INV-157, INV-159, and INV-161 are physiochemical combinations of lipoic acid and captopril. We sought to determine if these compounds have neuroprotective potential following middle cerebral artery occlusion (MCAO) in rats. Methods. Male Sprague-Dawley rats were injected intravenously with captopril (1–50 mg/kg) 30 minutes prior to MCAO. Blood pressure, heart rate, baroreceptor reflex sensitivity, and infarct size were measured. In addition, dose response effect on infarct size and cardiovascular parameters was determined using INV-155, INV-157, INV-159, and INV-161 and compared to captopril and LA. Results. Pretreatment with captopril and LA at all doses tested was neuroprotective. The compounds INV-159 (0.5–10 mg/kg) and INV-161 (1–10 mg/kg) produced a significant,dose-dependent decrease in infarct size. In contrast, INV-155 and INV-157 had no effect on infarct size. Conclusions. Combined pretreatment with captopril potentiated the neuroprotective benefit observed following LA alone. Both INV-159 and INV-161 were also neuroprotective. These results suggest that patients taking combinations of captopril and LA, either as combination therapy or in the form of INV-159 or INV-161, may also benefit from significant protection against cerebral infarction.
Collapse
|
13
|
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
|
14
|
Chung MH, Yang CC. Heart Rate Variability Across the Menstrual Cycle in Shift Work Nurses. ACTA ACUST UNITED AC 2011. [DOI: 10.1016/j.jecm.2011.04.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
15
|
Differential Neuroprotection of Selective Estrogen Receptor Agonists against Autonomic Dysfunction and Ischemic Cell Death in Permanent versus Reperfusion Injury. Adv Pharmacol Sci 2011; 2011:976951. [PMID: 21738528 PMCID: PMC3124232 DOI: 10.1155/2011/976951] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2011] [Revised: 03/03/2011] [Accepted: 03/04/2011] [Indexed: 11/17/2022] Open
Abstract
In the present study, we tested the hypothesis that selective activation of estrogen receptor subtypes (ERα and ERβ) would be neuroprotective following ischemia and/or ischemia-reperfusion, as well as prevent the associated autonomic dysfunction. The selective ERα agonist, PPT, when administered 30 min prior to occlusion of the middle cerebral artery (pMCAO), resulted in a dose-dependent neuroprotection as measured 6 hours postpermanent MCAO, but not following 30 mins of MCAO followed by 5.5 hrs of reperfusion (I/R). In contrast, 30 min pretreatment with the selective ERβ agonist, DPN, resulted in a dose-dependent neuroprotection following I/R, but was not protective following pMCAO. Both drugs prevented the ischemia-induced autonomic dysfunction as measured by a decrease in the baroreceptor reflex sensitivity (BRS). The data presented here suggest a differential role of each ER subtype in targeting the mechanisms of cell death that occur in ischemia versus reperfusion injury.
Collapse
|
16
|
Strom JO, Theodorsson E, Holm L, Theodorsson A. Different methods for administering 17beta-estradiol to ovariectomized rats result in opposite effects on ischemic brain damage. BMC Neurosci 2010; 11:39. [PMID: 20236508 PMCID: PMC2848231 DOI: 10.1186/1471-2202-11-39] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2009] [Accepted: 03/17/2010] [Indexed: 01/26/2023] Open
Abstract
Background Numerous stroke studies have controversially shown estrogens to be either neuroprotective or neurodamaging. The discordant results observed in rat brain ischemia models may be a consequence of discrepancies in estrogen administration modes resulting in plasma concentration profiles far from those intended. To test this hypothesis we reproduced in detail and extended an earlier study from our lab using a different mode of 17β-estradiol administration; home-made silastic capsules instead of commercial slow-release 17β-estradiol pellets. Four groups of female rats (n = 12) were ovariectomized and administered 17β-estradiol or placebo via silastic capsules. All animals underwent MCAo fourteen days after ovariectomy and were sacrificed three days later. Results In contrast to our earlier results using the commercial pellets, the group receiving 17β-estradiol during the entire experiment had significantly smaller lesions than the group receiving placebo (mean ± SEM: 3.85 ± 0.70% versus 7.15 ± 0.27% of total slice area, respectively; p = 0.015). No significant neuroprotection was found when the 17β-estradiol was administered only during the two weeks before or the three days immediately after MCAo. Conclusions The results indicate that different estrogen treatment regimens result in diametrically different effects on cerebral ischemia. Thus the effects of estrogens on ischemic damage seem to be concentration-related, with a biphasic, or even more complex, dose-response relation. These findings have implications for the design of animal experiments and also have a bearing on the estrogen doses used for peri-menopausal hormone replacement therapy.
Collapse
Affiliation(s)
- Jakob O Strom
- Institution of Clinical and Experimental Medicine/Department of Clinical Chemistry, Linkoping University, Linkoping, Sweden
| | | | | | | |
Collapse
|
17
|
Acute administration of non-classical estrogen receptor agonists attenuates ischemia-induced hippocampal neuron loss in middle-aged female rats. PLoS One 2010; 5:e8642. [PMID: 20062809 PMCID: PMC2799530 DOI: 10.1371/journal.pone.0008642] [Citation(s) in RCA: 122] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2009] [Accepted: 12/16/2009] [Indexed: 11/19/2022] Open
Abstract
Background Pretreatment with 17β-estradiol (E2) is profoundly neuroprotective in young animals subjected to focal and global ischemia. However, whether E2 retains its neuroprotective efficacy in aging animals, especially when administered after brain insult, is largely unknown. Methodology/Principal Findings We examined the neuroprotective effects of E2 and two agonists that bind to non-classical estrogen receptors, G1 and STX, when administered after ischemia in middle-aged rats after prolonged ovarian hormone withdrawal. Eight weeks after ovariectomy, middle-aged female rats underwent 10 minutes of global ischemia by four vessel occlusion. Immediately after reperfusion, animals received a single infusion of either E2 (2.25 µg), G1 (50 µg) or STX (50 µg) into the lateral ventricle (ICV) or a single systemic injection of E2 (100 µg/kg). Surviving pyramidal neurons in the hippocampal CA1 were quantified 1 week later. E2 and both agonists that target non-classical estrogen receptors (G1 and STX) administered ICV at the time of reperfusion provided significant levels of neuroprotection, with 55–60% of CA1 neurons surviving vs 15% survival in controls. A single systemic injection of a pharmacological dose of E2 also rescued approximately 50% of CA1 pyramidal neurons destined to die. To determine if E2 and G1 have similar mechanisms of action in hippocampal neurons, we compared the ability of E2 and G1 to modify CA1 pyramidal neuron responses to excitatory inputs from the Schaffer collaterals recorded in hippocampal slices derived from female rats not subjected to global ischemia. E2 and G1 (10 nM) significantly potentiated pyramidal neuron responses to excitatory inputs when applied to hippocampal slices. Conclusions/Significance These findings suggest (1) that middle-aged female rats retain their responsiveness to E2 even after a long period of hormone withdrawal, (2) that non-classical estrogen receptors may mediate the neuroprotective actions of E2 when given after ischemia, and (3) that the neuroprotective efficacy of estrogens may be related to their modulation of synaptic activity in hippocampal slices.
Collapse
|
18
|
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
|
19
|
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]
|
20
|
Abstract
1. In recent years, the role of oestrogen in women's health has been a subject of considerable scientific and popular debate. There is unquestionable evidence that oestrogen has both potent and long-lasting effects on several vital organ systems, including the cardiovascular system, the autonomic nervous system and, most recently, within the central nervous system itself. 2. The research and medical community continues to debate whether the benefits of oestrogen therapy outweigh the risks in the treatment of the symptoms of menopause, the attenuation of the risk for cardiovascular insults, such as stroke and heart disease, and even the retardation of the progression of Alzheimer's disease. 3. The recent evidence provided by the Heart and Estrogen/Progestin Replacement Study (HERS) II clinical trial suggesting that long-term exposure to combined oestrogen and progestin in post-menopausal women who have previously had a heart attack or stroke (for secondary prevention) may actually increase their risk of a subsequent cardiovascular insult has further fuelled the debate. However, there remain considerable gaps in our knowledge with respect to the actual mechanisms by which oestrogen exerts its various beneficial effects at the cellular level for the primary prevention of cardiovascular disease. This information is essential if we are to harness the positive aspects of oestrogen therapy in such a manner as to avoid or minimize the associated risks of increased oestrogen exposure in women who we know, with some certainty, to be at an increased risk of cancers of the uterus, cervix and breast tissue.
Collapse
Affiliation(s)
- T M Saleh
- Department of Biomedical Science, Atlantic Veterinary College and The Prince Edward Island Health Research Institute, University of Prince Edward Island, Charlottetown, Prince Edward Island, Canada.
| | | |
Collapse
|
21
|
Wilson BC, Connell B, Saleh TM. Relaxin-induced reduction of infarct size in male rats receiving MCAO is dependent on nitric oxide synthesis and not estrogenic mechanisms. Neurosci Lett 2006; 393:160-4. [PMID: 16233954 DOI: 10.1016/j.neulet.2005.09.059] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2005] [Revised: 09/14/2005] [Accepted: 09/23/2005] [Indexed: 10/25/2022]
Abstract
Relaxins are members of the insulin peptide superfamily. Previous evidence has shown that relaxin pretreatment reduces cortical infarct size in anesthetized, male rats receiving permanent middle cerebral artery occlusion (MCAO). Therefore, the current study was designed to determine if estrogenic mechanisms or nitric oxide production are involved in mediating this relaxin-induced neuroprotection. In separate groups of rats (n=4-6), the following drugs were injected directly into the cortex 30 min prior to MCAO: (a) relaxin, (b) relaxin and estrogen, and (c) relaxin and an estrogen receptor antagonist (ICI 182,780). To investigate the involvement of nitric oxide, relaxin or relaxin and an inhibitor of endothelial nitric oxide synthase (L-NIO) were injected i.v. 30 min prior to MCAO. Saline-treated rats (both intracortical (i.c.) and intravenously (i.v.)) served as controls. Brains were harvested 4h post stroke, coronally sectioned using a brain matrix and stained using 2,3,5-triphenoltetrazolium chloride (TTC). Digital photographs were taken of brain sections and the ratio comparing the area of the infarct to the area of the ipsilateral hemisphere was calculated. Mean ratios were compared using ANOVA and Tukey's test. Intracortical and intravenous relaxin pretreatment significantly reduced the infarct area in the cortex by 33.7 and 58.6%, respectively compared to saline-treated controls. This effect was not dependent on an interaction with estrogenic receptors as co-injection of relaxin and ICI 182,780 did not reverse this effect. However, inhibition of nitric oxide synthase significantly reduced the relaxin-mediated neuroprotection suggesting that relaxin may induce the endothelin-NOS cascade in cerebral vasculature causing vasodilation and improved perfusion of neural tissue.
Collapse
Affiliation(s)
- Brian C Wilson
- Department of Biology, Acadia University, Wolfville, NS, Canada B4P 2R6.
| | | | | |
Collapse
|
22
|
Wilson BC, Milne P, Saleh TM. Relaxin Pretreatment Decreases Infarct Size in Male Rats after Middle Cerebral Artery Occlusion. Ann N Y Acad Sci 2006; 1041:223-8. [PMID: 15956712 DOI: 10.1196/annals.1282.035] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
We studied the possible neuroprotective action of relaxin in a rat stroke model. Relaxin (10 ng in 200 nL saline) or saline was injected into the secondary somatosensory cortex of anesthetized rats. Thirty minutes after treatment, the right middle cerebral artery was occluded, causing ischemic conditions. Brains were removed 4 hours after stroke, and 1-mm coronal sections were stained using 2-3-5-triphenoltetrazolium chloride. Digital photographs were taken of the sections, and the ratio of infarct area to ipsilateral hemispheric area was calculated. Relaxin treatment significantly (P < .05) reduced this ratio compared with that of saline-treated controls. Results suggest that relaxin may prevent ischemia-induced cell death.
Collapse
Affiliation(s)
- Brian C Wilson
- Department of Biology, Acadia University, Wolfville, Nova Scotia B4P 2R6, Canada.
| | | | | |
Collapse
|
23
|
Goodenough S, Schleusner D, Pietrzik C, Skutella T, Behl C. Glycogen synthase kinase 3beta links neuroprotection by 17beta-estradiol to key Alzheimer processes. Neuroscience 2005; 132:581-9. [PMID: 15837120 DOI: 10.1016/j.neuroscience.2004.12.029] [Citation(s) in RCA: 105] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/21/2004] [Indexed: 01/26/2023]
Abstract
Estrogen exerts many of its receptor-mediated neuroprotective functions through the activation of various intracellular signal transduction pathways including the mitogen activating protein kinase (MAPK), phospho inositol-3 kinase and protein kinase C pathways. Here we have used a hippocampal slice culture model of kainic acid-induced neurotoxic cell death to show that estrogen can protect against oxidative cell death. We have previously shown that MAPK and glycogen synthase kinase-3beta (GSK-3beta) are involved in the cell death/cell survival induced by kainic acid. In this model and other cellular and in vivo models we have shown that estrogen can also cause the phosphorylation and hence inactivation of GSK-3beta, a known mediator of neuronal cell death. The effect of estrogen on GSK-3beta activity is estrogen receptor mediated. Further, this estrogen/GSK-3beta interaction may have functional consequences in cellular models of some key pathogenic pathways associated with Alzheimer's disease. More specifically, estrogen affects the basal levels of tau phosphorylation at a site known to be phosphorylated by GSK-3beta. Taken together, these data indicate a novel molecular and functional link between estrogen and GSK-3beta and may have implications for estrogen receptor modulation as a target for the prevention of neurodegenerative disorders.
Collapse
Affiliation(s)
- S Goodenough
- Institute for Physiological Chemistry and Pathobiochemistry, Johannes Gutenberg University, Medical School, Duesbergweg 6, D-55099 Mainz, Germany
| | | | | | | | | |
Collapse
|
24
|
Abstract
Women are protected from stroke relative to men until the years of menopause. Because stroke is the leading cause of serious, long-term disability in the United States, modeling sex-specific mechanisms and outcomes in animals is vital to research. Important research questions are focused on the effects of hormone replacement therapy, age, reproductive status, and identification of sex-specific risk factors. Available research relevant to stroke in the female has almost exclusively utilized rodent models. Gender-linked stroke outcomes are more detectable in experimental studies than in clinical trials and observational studies. Various estrogens have been extensively studied as neuroprotective agents in women, animals, and a variety of in vitro models of neural injury and degeneration. Most data in animal and cell models are based on 17 beta estradiol and suggest that this steroid is neuroprotective in injury from ischemia/reperfusion. However, current evidence for the clinical benefits of hormone replacement therapy is unclear. Future research in this area will need to expand into stroke models utilizing higher order, gyrencephalic animals such as nonhuman primates if we are to improve extrapolation to the human scenario and to direct and enhance the design of ongoing and future clinical studies and trials.
Collapse
Affiliation(s)
- Stephanie J Murphy
- Department of Anesthesiology and Peri-Operative Medicine, Oregon Health and Science University, Portland, OR, USA
| | | | | |
Collapse
|
25
|
Abstract
Estrogen replacement therapy enhances mood, delays cognitive decline, and reduces the risk of neurodegeneration. Our laboratory has shown previously that pretreatment with low physiological levels of estradiol protects against middle cerebral artery occlusion (MCAO)-induced brain injury during late phases of neuronal cell death. Immediate early genes (IEGs) are induced by various forms of brain injury, and their induction is known to be a critical step in programmed cell death. The current study tested the hypothesis that the ability of estradiol to reduce MCAO-induced cell death involves attenuation of expression of one or more IEGs. We examined the effects of MCAO on the temporospatial pattern of IEG expression and the modulation of this pattern by estradiol replacement. Rats were ovariectomized and treated with either vehicle or low physiological concentrations of estradiol. One week later, rats underwent MCAO and brains were collected 1, 4, 8, 16, and 24 hr later. We assessed IEG mRNAs in discrete regions of brain by RT-PCR at 24 hr. We examined expression of c-Fos mRNA and protein in greater detail using in situ hybridization and immunohistochemistry to delineate the time course and specific regions of cortex in which estradiol influenced its expression. Our results reveal that c-fos, fosB, c-jun, and junB levels were upregulated at 24 hr. Furthermore, estradiol selectively affected the expression of c-Fos mRNA and protein by attenuating the injury-induced increase in a time- and region-specific manner. Our findings strongly suggest that the ability of estradiol to protect against MCAO-induced cell death involves attenuation of c-Fos induction.
Collapse
|
26
|
Liu CC, Kuo TBJ, Yang CCH. Effects of estrogen on gender-related autonomic differences in humans. Am J Physiol Heart Circ Physiol 2003; 285:H2188-93. [PMID: 12881217 DOI: 10.1152/ajpheart.00256.2003] [Citation(s) in RCA: 158] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Our previous studies demonstrated that premenopausal women have dominant vagal and subordinate sympathetic activity compared with age-matched men. This study was designed to investigate the role of estrogen in gender-related autonomic differences. We evaluated the heart rate variability of four healthy groups: age-matched postmenopausal women without hormone replacement therapy (PM), postmenopausal women on conjugated estrogen replacement therapy (PME), men, and non-age-matched premenopausal women (PreM). Frequency-domain analysis of short-term and stationary R-R intervals was performed to evaluate low-frequency power (LF; 0.04-0.15 Hz), high-frequency power (HF; 0.15-0.40 Hz), the ratio of LF to HF (LF/HF), and LF in normalized units (LF%). No gender-related autonomic differences existed between the PM and men groups, but they did exist between the PME and men group. Compared with the PreM group, the PM group had a lower HF and higher LF% and LF/HF. Compared with the PM group, the PME group had a higher HF but lower LF% and LF/HF. These results suggest that conjugated estrogen replacement therapy may facilitate vagal and attenuate sympathetic regulation of heart rate in postmenopausal women. In addition, estrogen may play an important role in gender-related autonomic differences.
Collapse
Affiliation(s)
- C C Liu
- Shou-Feng Health Station, Hualien 974, Taiwan
| | | | | |
Collapse
|
27
|
Murphy S, McCullough L, Littleton-Kearney M, Hurn P. Estrogen and selective estrogen receptor modulators: neuroprotection in the Women's Health Initiative era. Endocrine 2003; 21:17-26. [PMID: 12777699 DOI: 10.1385/endo:21:1:17] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2003] [Revised: 02/04/2003] [Accepted: 02/11/2003] [Indexed: 11/11/2022]
Abstract
Estrogen has been comprehensively studied as a neuroprotective agent in women, animals, and a variety of in vitro models of neural injury and degeneration. Most data suggest that estrogen can benefit the ischemic brain and reduce cell death. However, recent data from the Women's Health Initiative have raised concerns about the utility and safety of chronic estrogen use in women. While estrogen is a potent and reproducible neuroprotectant in animals and in vitro, its current administration in women has had unanticipated and paradoxical effects. Nonetheless, estrogen's diverse actions make it an ideal prototype for developing new neuroprotectants such as selective estrogen receptor modulators (SERMs). SERMs represent a class of drugs with mixed estrogen agonistic and antagonistic activity. Experimental and clinical data suggest a neuroprotective role for SERMs in normal and injured brain. The discrepancy among observational studies, preclinical data, and clinical trials emphasizes the need for further study of the mechanisms leading to the increased incidence of stroke observed in postmenopausal women. Research is still needed to optimize combined or estrogen alone hormone replacement therapy options as well as the prevention/management of cerebrovascular/ central nervous system disorders. This review critiques estrogen and SERMs' neuroprotective potential in experimental and clinical studies of stroke and cerebrovascular disease.
Collapse
Affiliation(s)
- Stephanie Murphy
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins Medical Institutions, Baltimore, MD, USA.
| | | | | | | |
Collapse
|
28
|
Abstract
In addition to its role as a sex hormone, oestrogen affects the structure and function of the nervous system. Oestrogen receptors are expressed in brain regions that are involved in sex differentiation and maturation. But in addition to its well-known effects, oestrogen also has important neuroprotective actions that are both dependent and independent of a nuclear oestrogen-receptor activity. Furthermore, oestrogen can interact with neuroprotective intracellular signalling pathways and is itself a neuroprotective antioxidant. Understanding the mechanisms of oestrogen action will be crucial to determine its potential as a therapeutic agent, particularly in the elderly.
Collapse
Affiliation(s)
- Christian Behl
- Max Planck Institute of Psychiatry, 80804 Munich, Germany.
| |
Collapse
|
29
|
Saleh TM, Cribb AE, Connell BJ. Reduction in infarct size by local estrogen does not prevent autonomic dysfunction after stroke. Am J Physiol Regul Integr Comp Physiol 2001; 281:R2088-95. [PMID: 11705796 DOI: 10.1152/ajpregu.2001.281.6.r2088] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Systemic estrogen administration in male rats has been shown to normalize the autonomic dysfunction and reduce the infarct size after permanent middle cerebral artery occlusion (MCAO). Therefore, the present investigation determined if local microinjection of estrogen at the site of the infarct also promoted recovery of autonomic function and reduction of the infarct size. Experiments were done in anesthetized (thiobutabarbitol sodium; 100 mg/kg) male Sprague-Dawley rats instrumented to record baseline and reflex changes in cardiovascular and autonomic parameters. The right middle cerebral artery was permanently occluded using bipolar coagulation. Local microinjection of estrogen into the insular cortex before MCAO significantly reduced the infarct size but did not attenuate the MCAO-induced autonomic dysfunction. Injection of ICI-182,780 alone significantly increased infarct area; however, the greater infarct area was not associated with enhanced autonomic dysfunction. These results suggest that within the insula, endogenous estrogen activity can affect the extent of MCAO-induced cell death, but extracortical central nervous system sites may be responsible for mediating the beneficial effects of estrogen on the autonomic disturbances.
Collapse
Affiliation(s)
- T M Saleh
- Department of Anatomy and Physiology, Atlantic Veterinary College, Univ. of Prince Edward Island, Charlottetown, Prince Edward Island, Canada C1A 4P3.
| | | | | |
Collapse
|