1
|
Pradhyumnan H, Patel SH, Furones-Alonso O, Zhao W, Bramlett HM, Raval AP. Electronic Cigarette Vape Exposure Exacerbates Post-Ischemic Outcomes in Female but Not in Male Rats. Stroke 2024; 55:735-746. [PMID: 38323450 PMCID: PMC10940219 DOI: 10.1161/strokeaha.123.046101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 01/11/2024] [Indexed: 02/08/2024]
Abstract
BACKGROUND Nicotine-containing electronic cigarette (EC) vaping has become popular worldwide, and our understanding of the effects of vaping on stroke outcomes is elusive. Using a rat model of transient middle cerebral artery occlusion, the current exploratory study aims to evaluate the sex-dependent effects of EC exposure on brain energy metabolism and stroke outcomes. METHODS Adult Sprague-Dawley rats of both sexes were randomly assigned to air/EC vapor (5% nicotine Juul pods) exposure for 16 nights, followed by randomization into 3 cohorts. The first cohort underwent exposure to air/EC preceding randomization to transient middle cerebral artery occlusion (90 minutes) or sham surgery, followed by survival for 21 days. During the survival period, rats underwent sensorimotor and Morris water maze testing. Subsequently, brains were collected for histopathology. A second cohort was exposed to air/EC after which brains were collected for unbiased metabolomics analysis. The third cohort of animals was exposed to air/EC and received transient middle cerebral artery occlusion/sham surgery, and brain tissue was collected 24 hours later for biochemical analysis. RESULTS In females, EC significantly increased (P<0.05) infarct volumes by 94% as compared with air-exposed rats, 165±50 mm3 in EC-exposed rats, and 85±29 mm3 in air-exposed rats, respectively, while in males such a difference was not apparent. Morris water maze data showed significant deficits in spatial learning and working memory in the EC sham or transient middle cerebral artery occlusion groups compared with the respective air groups in rats of both sexes (P<0.05). Thirty-two metabolites of carbohydrate, glycolysis, tricarboxylic acid cycle, and lipid metabolism were significantly altered (P≤0.05) due to EC, 23 of which were specific for females. Steady-state protein levels of hexokinase significantly decreased (P<0.05) in EC-exposed females; however, these changes were not seen in males. CONCLUSIONS Even brief EC exposure over 2 weeks impacts brain energy metabolism, exacerbates infarction, and worsens poststroke cognitive deficits in working memory more in female than male rats.
Collapse
Affiliation(s)
- Hari Pradhyumnan
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Shahil H. Patel
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Ofelia Furones-Alonso
- Department of Neurological Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Weizhao Zhao
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL 33146, USA
| | - Helen M. Bramlett
- Department of Neurological Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- Neuroscience Program, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- Bruce W. Carter Department of Veterans Affairs Medical Center, Miami, FL 33136, USA
| | - Ami P. Raval
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- Neuroscience Program, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- Bruce W. Carter Department of Veterans Affairs Medical Center, Miami, FL 33136, USA
| |
Collapse
|
2
|
Pradhyumnan H, Perez GG, Patel SH, Blaya MO, Bramlett HM, Raval AP. A Perspective on Hormonal Contraception Usage in Central Nervous System Injury. J Neurotrauma 2024; 41:541-551. [PMID: 37975282 DOI: 10.1089/neu.2023.0219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2023] Open
Abstract
Naturally occurring life stages in women are associated with changes in the milieu of endogenous ovarian hormones. Women of childbearing age may be exposed to exogenous ovarian hormone(s) because of their use of varying combinations of estrogen and progesterone hormones-containing oral contraceptives (OC; also known as "the pill"). If women have central nervous system (CNS) injury such as spinal cord injury (SCI) and traumatic brain injury (TBI) during their childbearing age, they are likely to retain their reproductive capabilities and may use OC. Many deleterious side effects of long-term OC use have been reported, such as aberrant blood clotting and endothelial dysfunction that consequently increase the risk of myocardial infarction, venous thromboembolism, and ischemic brain injury. Although controversial, studies have suggested that OC use is associated with neuropsychiatric ramifications, including uncontrollable mood swings and poorer cognitive performance. Our understanding about how the combination of endogenous hormones and OC-conferred exogenous hormones affect outcomes after CNS injuries remains limited. Therefore, understanding the impact of OC use on CNS injury outcomes needs further investigation to reveal underlying mechanisms, promote reporting in clinical or epidemiological studies, and raise awareness of possible compounded consequences. The goal of the current review is to discuss the impacts of CNS injury on endogenous ovarian hormones and vice-versa, as well as the putative consequences of exogenous ovarian hormones (OC) on the CNS to identify potential gaps in our knowledge to consider for future laboratory, epidemiological, and clinical studies.
Collapse
Affiliation(s)
- Hari Pradhyumnan
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratory (CVDRL), Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Gina G Perez
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratory (CVDRL), Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Shahil H Patel
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratory (CVDRL), Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Meghan O Blaya
- Department of Neurological Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida, USA
- The Miami Project to Cure Paralysis, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Helen M Bramlett
- Department of Neurological Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida, USA
- The Miami Project to Cure Paralysis, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida, USA
- Bruce W. Carter Department of Veterans Affairs Medical Center, Miami, Florida, USA
| | - Ami P Raval
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratory (CVDRL), Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida, USA
- Bruce W. Carter Department of Veterans Affairs Medical Center, Miami, Florida, USA
| |
Collapse
|
3
|
Contreras CM, Gutiérrez-García AG. Prelimbic and infralimbic responsivity to amygdala input is modified by gonadal hormones in parallel to low anxiety-like behavior in ovariectomized rats. Behav Brain Res 2024; 459:114795. [PMID: 38048910 DOI: 10.1016/j.bbr.2023.114795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 11/29/2023] [Accepted: 11/30/2023] [Indexed: 12/06/2023]
Abstract
Gonadal hormones may influence sexual activity by reducing anxiety. The basolateral amygdala (BLA) and prelimbic (PL) and infralimbic (IL) cortical regions comprise a loop that is related to fear, anxiety, and social behavior. In female ovariectomized rats, actions of estradiol, progesterone, and sequential estradiol and progesterone administration were explored in the open field test (OFT) and plus maze test (PMT) to evaluate signs of anxiety-like behavior. The three hormonal treatments reduced indicators of anxiety in the PMT but did not influence behavior in the OFT. In the same behaviorally tested rats under urethane anesthesia, single-unit extracellular recordings were obtained from the PL and IL during electrical stimulation of the BLA. The analysis of 250 ms peristimulus histograms showed that BLA stimulation produced two kinds of response. A small group of neurons increased their firing rate after BLA stimulation. Most neurons exhibited a reduction of spiking. Neurons that increased their firing rate after BLA stimulation did not show any difference with the hormonal treatments. In neurons that were inhibited by BLA stimulation, estradiol reduced the neuronal firing rate in the PL and IL, and progesterone alone and the sequential administration of estradiol followed by progesterone administration 24 h later (priming) increased the firing rate during the 240 ms before BLA stimulation. Analyses of responsivity of the PL and IL during electrical stimulation of the BLA indicated that estradiol, progesterone, and estradiol followed by progesterone administration 24 h later (priming) reduced inhibitory actions of the BLA on the PL but not IL. In the BLA-IL connection, progesterone exacerbated the inhibitory response. These findings indicate that anxiolytic actions of estradiol, progesterone, and estradiol followed by progesterone administration 24 h later (priming) correspond to lower BLA-PL responsivity. Actions of progesterone on BLA-IL responsivity appear to contribute to sexual activity by interacting with other forebrain structures that are also related to sexual receptivity.
Collapse
Affiliation(s)
- Carlos M Contreras
- Unidad Periférica-Xalapa, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Xalapa, Veracruz, Mexico.
| | - Ana G Gutiérrez-García
- Laboratorio de Neurofarmacología, Instituto de Neuroetología, Universidad Veracruzana, Xalapa, Veracruz, Mexico
| |
Collapse
|
4
|
Escobar I, Xu J, Jackson CW, Stegelmann SD, Fagerli EA, Dave KR, Perez-Pinzon MA. Resveratrol Preconditioning Protects Against Ischemia-Induced Synaptic Dysfunction and Cofilin Hyperactivation in the Mouse Hippocampal Slice. Neurotherapeutics 2023; 20:1177-1197. [PMID: 37208551 PMCID: PMC10457274 DOI: 10.1007/s13311-023-01386-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/23/2023] [Indexed: 05/21/2023] Open
Abstract
Perturbations in synaptic function are major determinants of several neurological diseases and have been associated with cognitive impairments after cerebral ischemia (CI). Although the mechanisms underlying CI-induced synaptic dysfunction have not been well defined, evidence suggests that early hyperactivation of the actin-binding protein, cofilin, plays a role. Given that synaptic impairments manifest shortly after CI, prophylactic strategies may offer a better approach to prevent/mitigate synaptic damage following an ischemic event. Our laboratory has previously demonstrated that resveratrol preconditioning (RPC) promotes cerebral ischemic tolerance, with many groups highlighting beneficial effects of resveratrol treatment on synaptic and cognitive function in other neurological conditions. Herein, we hypothesized that RPC would mitigate hippocampal synaptic dysfunction and pathological cofilin hyperactivation in an ex vivo model of ischemia. Various electrophysiological parameters and synaptic-related protein expression changes were measured under normal and ischemic conditions utilizing acute hippocampal slices derived from adult male mice treated with resveratrol (10 mg/kg) or vehicle 48 h prior. Remarkably, RPC significantly increased the latency to anoxic depolarization, decreased cytosolic calcium accumulation, prevented aberrant increases in synaptic transmission, and rescued deficits in long-term potentiation following ischemia. Additionally, RPC upregulated the expression of the activity-regulated cytoskeleton associated protein, Arc, which was partially required for RPC-mediated attenuation of cofilin hyperactivation. Taken together, these findings support a role for RPC in mitigating CI-induced excitotoxicity, synaptic dysfunction, and pathological over-activation of cofilin. Our study provides further insight into mechanisms underlying RPC-mediated neuroprotection against CI and implicates RPC as a promising strategy to preserve synaptic function after ischemia.
Collapse
Affiliation(s)
- Iris Escobar
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, PO Box 016960, Miami, FL, 33101, USA
- Department of Neurology, University of Miami Leonard M. Miller School of Medicine, PO Box 016960, Miami, FL, 33101, USA
- Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, PO Box 016960, Miami, FL, 33101, USA
| | - Jing Xu
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, PO Box 016960, Miami, FL, 33101, USA
- Department of Neurology, University of Miami Leonard M. Miller School of Medicine, PO Box 016960, Miami, FL, 33101, USA
- Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, PO Box 016960, Miami, FL, 33101, USA
| | - Charles W Jackson
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, PO Box 016960, Miami, FL, 33101, USA
- Department of Neurology, University of Miami Leonard M. Miller School of Medicine, PO Box 016960, Miami, FL, 33101, USA
- Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, PO Box 016960, Miami, FL, 33101, USA
| | - Samuel D Stegelmann
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, PO Box 016960, Miami, FL, 33101, USA
- Department of Neurology, University of Miami Leonard M. Miller School of Medicine, PO Box 016960, Miami, FL, 33101, USA
| | - Eric A Fagerli
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, PO Box 016960, Miami, FL, 33101, USA
- Department of Neurology, University of Miami Leonard M. Miller School of Medicine, PO Box 016960, Miami, FL, 33101, USA
- Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, PO Box 016960, Miami, FL, 33101, USA
| | - Kunjan R Dave
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, PO Box 016960, Miami, FL, 33101, USA
- Department of Neurology, University of Miami Leonard M. Miller School of Medicine, PO Box 016960, Miami, FL, 33101, USA
- Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, PO Box 016960, Miami, FL, 33101, USA
| | - Miguel A Perez-Pinzon
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, PO Box 016960, Miami, FL, 33101, USA.
- Department of Neurology, University of Miami Leonard M. Miller School of Medicine, PO Box 016960, Miami, FL, 33101, USA.
- Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, PO Box 016960, Miami, FL, 33101, USA.
| |
Collapse
|
5
|
Pradhyumnan H, Reddy V, Bassett ZQ, Patel SH, Zhao W, Dave KR, Perez-Pinzon MA, Bramlett HM, Raval AP. Post-stroke periodic estrogen receptor-beta agonist improves cognition in aged female rats. Neurochem Int 2023; 165:105521. [PMID: 36933865 DOI: 10.1016/j.neuint.2023.105521] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 03/14/2023] [Accepted: 03/15/2023] [Indexed: 03/18/2023]
Abstract
Women have a higher risk of having an ischemic stroke and increased cognitive decline after stroke as compared to men. The female sex hormone 17β-estradiol (E2) is a potent neuro- and cognitive-protective agent. Periodic E2 or estrogen receptor subtype-beta (ER-β) agonist pre-treatments every 48 h before an ischemic episode ameliorated ischemic brain damage in young ovariectomized or reproductively senescent (RS) aged female rats. The current study aims to investigate the efficacy of post-stroke ER-β agonist treatments in reducing ischemic brain damage and cognitive deficits in RS female rats. Retired breeder (9-10 months) Sprague-Dawley female rats were considered RS after remaining in constant diestrus phase for more than a month. The RS rats were exposed to transient middle cerebral artery occlusion (tMCAO) for 90 min and treated with either ER-β agonist (beta 2, 3-bis(4-hydroxyphenyl) propionitrile; DPN; 1 mg/kg; s.c.) or DMSO vehicle at 4.5 h after induction of tMCAO. Subsequently, rats were treated with either ER-β agonist or DMSO vehicle every 48 h for ten injections. Forty-eight hours after the last treatment, animals were tested for contextual fear conditioning to measure post-stroke cognitive outcome. Neurobehavioral testing, infarct volume quantification, and hippocampal neuronal survival were employed to determine severity of stroke. Periodic post-stroke ER-β agonist treatment reduced infarct volume, improved recovery of cognitive capacity by increasing freezing in contextual fear conditioning, and decreased hippocampal neuronal death in RS female rats. These data suggest that periodic post-stroke ER-β agonist treatment to reduce stroke severity and improve post-stroke cognitive outcome in menopausal women has potential for future clinical investigation.
Collapse
Affiliation(s)
- Hari Pradhyumnan
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, 33136, USA; Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, 33136, USA
| | - Varun Reddy
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, 33136, USA; Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, 33136, USA
| | - Zoe Q Bassett
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, 33136, USA; Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, 33136, USA
| | - Shahil H Patel
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, 33136, USA; Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, 33136, USA
| | - Weizhao Zhao
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL, 33146, USA
| | - Kunjan R Dave
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, 33136, USA; Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, 33136, USA; Neuroscience Program, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, 33136, USA
| | - Miguel A Perez-Pinzon
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, 33136, USA; Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, 33136, USA; Neuroscience Program, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, 33136, USA
| | - Helen M Bramlett
- Department of Neurological Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, 33136, USA; Neuroscience Program, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, 33136, USA; Bruce W. Carter Department of Veterans Affairs Medical Center, Miami, FL, 33136, USA
| | - Ami P Raval
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, 33136, USA; Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, 33136, USA; Neuroscience Program, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, 33136, USA; Bruce W. Carter Department of Veterans Affairs Medical Center, Miami, FL, 33136, USA.
| |
Collapse
|
6
|
Patel SH, Timón-Gómez A, Pradhyumnan H, Mankaliye B, Dave KR, Perez-Pinzon MA, Raval AP. The Impact of Nicotine along with Oral Contraceptive Exposure on Brain Fatty Acid Metabolism in Female Rats. Int J Mol Sci 2022; 23:ijms232416075. [PMID: 36555717 PMCID: PMC9780830 DOI: 10.3390/ijms232416075] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 12/06/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
Smoking-derived nicotine (N) and oral contraceptive (OC) synergistically exacerbate ischemic brain damage in females, and the underlying mechanisms remain elusive. In a previous study, we showed that N + OC exposure altered brain glucose metabolism in females. Since lipid metabolism complements glycolysis, the current study aims to examine the metabolic fingerprint of fatty acids in the brain of female rats exposed to N+/-OC. Adolescent and adult Sprague-Dawley female rats were randomly (n = 8 per group) exposed to either saline or N (4.5 mg/kg) +/-OC (combined OC or placebo delivered via oral gavage) for 16-21 days. Following exposure, brain tissue was harvested for unbiased metabolomic analysis (performed by Metabolon Inc., Morrisville, NC, USA) and the metabolomic profile changes were complemented with Western blot analysis of key enzymes in the lipid pathway. Metabolomic data showed significant accumulation of fatty acids and phosphatidylcholine (PC) metabolites in the brain. Adolescent, more so than adult females, exposed to N + OC showed significant increases in carnitine-conjugated fatty acid metabolites compared to saline control animals. These changes in fatty acyl carnitines were accompanied by an increase in a subset of free fatty acids, suggesting elevated fatty acid β-oxidation in the mitochondria to meet energy demand. In support, β-hydroxybutyrate was significantly lower in N + OC exposure groups in adolescent animals, implying a complete shunting of acetyl CoA for energy production via the TCA cycle. The reported changes in fatty acids and PC metabolism due to N + OC could inhibit post-translational palmitoylation of membrane proteins and synaptic vesicle formation, respectively, thus exacerbating ischemic brain damage in female rats.
Collapse
Affiliation(s)
- Shahil H. Patel
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Alba Timón-Gómez
- Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Hari Pradhyumnan
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Berk Mankaliye
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Kunjan R. Dave
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- Neuroscience Program, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Miguel A. Perez-Pinzon
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- Neuroscience Program, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Ami P. Raval
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- Neuroscience Program, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- Bruce W. Carter Department of Veterans Affairs Medical Center, Miami, FL 33136, USA
- Correspondence: ; Tel.: +1-(305)-243-7491; Fax: +1-(305)-243-6955
| |
Collapse
|
7
|
Red Cell Microparticles Suppress Hematoma Growth Following Intracerebral Hemorrhage in Chronic Nicotine-Exposed Rats. Int J Mol Sci 2022; 23:ijms232315167. [PMID: 36499494 PMCID: PMC9736308 DOI: 10.3390/ijms232315167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/21/2022] [Accepted: 11/22/2022] [Indexed: 12/03/2022] Open
Abstract
Spontaneous intracerebral hemorrhage (sICH) is a disabling stroke sub-type, and tobacco use is a prominent risk factor for sICH. We showed that chronic nicotine exposure enhances bleeding post-sICH. Reduction of hematoma growth is a promising effective therapy for sICH in smoking subjects. Red-blood-cell-derived microparticles (RMPs) are hemostatic agents that limit hematoma expansion following sICH in naïve rats. Considering the importance of testing the efficacy of experimental drugs in animal models with a risk factor for a disease, we tested RMP efficacy and the therapeutic time window in limiting hematoma growth post-sICH in rats exposed to nicotine. Young rats were chronically treated with nicotine using osmotic pumps. sICH was induced in rats using an injection of collagenase in the right striatum. Vehicle/RMPs were administered intravenously. Hematoma volume and neurological impairment were quantified ≈24 h after sICH. Hematoma volumes in male and female nicotine-exposed rats that were treated with RMPs at 2 h post-sICH were significantly lower by 26 and 31% when compared to their respective control groups. RMP therapy was able to limit hematoma volume when administered up to 4.5 h post-sICH in animals of both sexes. Therefore, RMPs may limit hematoma growth in sICH patients exposed to tobacco use.
Collapse
|
8
|
Introduction to the special issue on neurological disorders across the female life span. Neurobiol Dis 2022; 174:105886. [DOI: 10.1016/j.nbd.2022.105886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
9
|
Kerr N, Sanchez J, Moreno WJ, Furones-Alonso OE, Dietrich WD, Bramlett HM, Raval AP. Post-stroke low-frequency whole-body vibration improves cognition in middle-aged rats of both sexes. Front Aging Neurosci 2022; 14:942717. [PMID: 36062148 PMCID: PMC9428155 DOI: 10.3389/fnagi.2022.942717] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 07/26/2022] [Indexed: 11/18/2022] Open
Abstract
Low-frequency whole-body vibration (WBV; 40 Hz), a low impact form of exercise, intervention for a month following moderate transient middle-cerebral artery occlusion (tMCAO) reduces infarct volume and improves motor function in reproductively senescent, middle-aged female rats. Since post-stroke cognitive decline remains a significant problem, the current study aims to investigate the efficacy of WBV in ameliorating post-tMCAO cognitive deficits and to determine the underlying putative mechanism(s) conferring benefits of WBV in middle-aged rats. Middle-aged rats of both sexes were randomly assigned to tMCAO (90 min) or sham surgery followed by exposure to either WBV (twice a day for 15 min each for 5 days a week over a month) or no WBV treatment groups. Following the last WBV treatment, rats were tested for hippocampus-dependent learning and memory using a water maze followed by harvesting brain and blood samples for histopathological and inflammatory marker analyses, respectively. Results show that post-tMCAO WBV significantly lessens cognitive deficits in rats of both sexes. Post-tMCAO WBV significantly decreased circulating pro-inflammatory cytokines and increased serum levels of irisin, a muscle-derived hormone that may play a role in brain metabolism and inflammation regulation, which suggests putative beneficial mechanisms of WBV.
Collapse
Affiliation(s)
- Nadine Kerr
- Department of Neurological Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Juliana Sanchez
- Department of Neurological Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, United States
| | - William Javier Moreno
- Department of Neurological Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Ofelia E. Furones-Alonso
- Department of Neurological Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, United States
| | - W. Dalton Dietrich
- Department of Neurological Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Helen M. Bramlett
- Department of Neurological Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, United States
- Bruce W. Carter Department of Veterans Affairs Medical Center, Miami, FL, United States
- *Correspondence: Helen M. Bramlett,
| | - Ami P. Raval
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratory, Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, United States
- Ami P. Raval,
| |
Collapse
|
10
|
Chronic Nicotine Exposure Increases Hematoma Expansion Following Collagenase-Induced Intracerebral Hemorrhage in Rats. Biomolecules 2022; 12:biom12050621. [PMID: 35625548 PMCID: PMC9138464 DOI: 10.3390/biom12050621] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 04/17/2022] [Accepted: 04/18/2022] [Indexed: 02/04/2023] Open
Abstract
Spontaneous intracerebral hemorrhage (sICH) is a deadly stroke subtype, and tobacco use increases sICH risk. However epidemiological studies show that, there are no confirmatory studies showing the effect of tobacco use on sICH outcome. Therefore, we evaluated the effect of chronic nicotine exposure (as a surrogate for tobacco use) on outcomes following sICH. Young male and female rats were randomly assigned to either nicotine (4.5 mg/kg b.w. per day) or vehicle (saline) treatment (2–3 weeks) groups. sICH was induced by injecting collagenase into the right striatum. Neurological score and hematoma volume were determined 24 h post-sICH. The hematoma volumes in nicotine-treated male and female rats were significantly higher by 42% and 48% when compared to vehicle-treated male and female rats, respectively. Neurological deficits measured in terms of neurological score for the nicotine-treated male and female groups were significantly higher when compared to the respective vehicle-treated male and female groups. Our results show that chronic nicotine exposure increases hematoma volume post-sICH in rats of both sexes. Identifying the mechanism of nicotine-dependent increase in hematoma growth post-sICH will be crucial to understanding the detrimental effect of tobacco use on the severity of bleeding following intracerebral hemorrhage.
Collapse
|
11
|
Blaya MO, Raval AP, Bramlett HM. Traumatic brain injury in women across lifespan. Neurobiol Dis 2022; 164:105613. [PMID: 34995753 DOI: 10.1016/j.nbd.2022.105613] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 12/16/2021] [Accepted: 01/03/2022] [Indexed: 11/27/2022] Open
Abstract
Traumatic brain injury (TBI) is a leading cause of death and disability and a global public health challenge. Every year more than 50 million people suffer a TBI, and it is estimated that 50% of the global population will experience at least one TBI in their lifetime. TBI affects both men and women of all ages, however there is a male bias in TBI research as women have frequently been left out of the literature despite irrefutable evidence of male and female dimorphism in several posttraumatic measures. Women uniquely experience distinct life stages marked by levels of endogenous circulating sex hormones, as well as by physiological changes that are nonexistent in men. In addition to generalized sex-specific differences, a woman's susceptibility, neurological outcomes, and treatment success may vary considerably depending upon when in her lifespan she incurred a traumatic insult. How women impacted by TBI might differ from other women as a factor of age and physiology is not well understood. Furthermore, there is a gap in the knowledge of what happens when TBI occurs in the presence of certain sex-specific and sex-nonspecific variables, such as during pregnancy, with oral contraceptive use, in athletics, in cases of addiction and nicotine consumption, during perimenopause, postmenopause, in frailty, among others. Parsing out how hormone-dependent and hormone-independent lifespan variables may influence physiological, neurodegenerative, and functional outcomes will greatly contribute to future investigative studies and direct therapeutic strategies. The goal of this review is to aggregate the knowledge of prevalence, prognosis, comorbid risk, and response of women incurring TBI at differing phases of lifespan. We strive to illuminate commonalities and disparities among female populations, and to pose important questions to highlight gaps in the field in order to further the endeavor of targeted treatment interventions in a patient-specific manner.
Collapse
Affiliation(s)
- Meghan O Blaya
- Department of Neurological Surgery, University of Miami, Miami, Florida, USA; The Miami Project to Cure Paralysis, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Ami P Raval
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratory, Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Helen M Bramlett
- Department of Neurological Surgery, University of Miami, Miami, Florida, USA; The Miami Project to Cure Paralysis, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida, USA; Bruce W. Carter Department of Veterans Affairs Medical Center, Miami, Florida, USA.
| |
Collapse
|
12
|
d'Adesky N, Diaz F, Zhao W, Bramlett HM, Perez-Pinzon MA, Dave KR, Raval AP. Nicotine Exposure Along with Oral Contraceptive Treatment in Female Rats Exacerbates Post-cerebral Ischemic Hypoperfusion Potentially via Altered Histamine Metabolism. Transl Stroke Res 2021; 12:817-828. [PMID: 33130995 DOI: 10.1007/s12975-020-00854-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 09/19/2020] [Accepted: 09/23/2020] [Indexed: 10/23/2022]
Abstract
Smoking-derived nicotine (N) and oral contraceptives (OCs) synergistically exacerbate ischemic brain damage in the female, and the underlying mechanisms remain elusive. Our published study showed that N toxicity is exacerbated by OC via altered mitochondrial electron transport chain function. Because mitochondria play an important role in cellular metabolism, we investigated the global metabolomic profile of brains of adolescent and adult female Sprague-Dawley rats exposed to N with or without OC (N+/-OC). Rats were randomly exposed to saline or N+/-OC for 16-21 days followed by random allocation into two cohorts. The first cohort was used to characterize the cortical metabolome. Pathway enrichment analysis showed a significant increase in several histamine metabolites including 1-methylhistamine, 1-methyl-4-imidazoleacetate, and 1-ribosyl-imidazleacetate, along with carnosine and homocarnosine in adolescent and adult animals treated with N and N+OC in relation to respective saline controls, which may be reflective of altered histamine metabolism with nicotine treatment. We also observed reduced levels of the neurotransmitters N-acetyl-aspartyl-glutamate (NAAG), gamma-aminobutyrate (GABA), and N-methyl-GABA in N+OC treatment in adolescent animals. The second cohort underwent bilateral carotid artery occlusion and hypotension followed by cerebral blood flow (CBF) assessment a day later. Autoradiographic images of the brain 24 h after ischemic episodes showed severe reduction in cortical and hippocampal local CBF in N+/-OC-exposed rats compared with saline treated. Because GABA and histamine are critical for CBF maintenance, altered metabolism of these neurotransmitters may be responsible for observed severe post-ischemic hypoperfusion, which in turn exacerbates ischemic brain damage.
Collapse
Affiliation(s)
- Nathan d'Adesky
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratory, Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, 1420 NW 9th Avenue, Neurology Research Building, Room # 203H, Miami, FL, 33136, USA
| | - Francisca Diaz
- Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Weizhao Zhao
- Biomedical Engineering, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Helen M Bramlett
- Department of Neurological Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
- Bruce W. Carter Department of Veterans Affairs Medical Center, Miami, FL, USA
| | - Miguel A Perez-Pinzon
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratory, Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, 1420 NW 9th Avenue, Neurology Research Building, Room # 203H, Miami, FL, 33136, USA
| | - Kunjan R Dave
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratory, Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, 1420 NW 9th Avenue, Neurology Research Building, Room # 203H, Miami, FL, 33136, USA
| | - Ami P Raval
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratory, Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, 1420 NW 9th Avenue, Neurology Research Building, Room # 203H, Miami, FL, 33136, USA.
| |
Collapse
|
13
|
Diaz F, Raval AP. Simultaneous nicotine and oral contraceptive exposure alters brain energy metabolism and exacerbates ischemic stroke injury in female rats. J Cereb Blood Flow Metab 2021; 41:793-804. [PMID: 32538281 PMCID: PMC7983508 DOI: 10.1177/0271678x20925164] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Smoking-derived nicotine (N) and oral contraceptives (OC) synergistically exacerbate ischemic brain damage in the females and underlying mechanisms remain elusive. Our published study showed that N toxicity is exacerbated by OC via altered mitochondrial function owing to a defect in the activity of cytochrome c oxidase. Here, we investigated the global metabolomic profile of brains of adolescent female Sprague-Dawley rats exposed to N ± OC. Rats were randomly exposed to saline or N + /-OC for 16-21 days followed by random allocation into two cohorts. One cohort underwent transient middle cerebral artery occlusion and histopathology was performed 30 days later. From the second cohort, cortical tissues were collected for an unbiased global metabolomic profile. Pathway enrichment analysis showed significant decrease in glucose, glucose 6-phosphate and fructose-6-phosphate, along with a significant increase in pyruvate in the N + /-OC exposed groups when compared to saline (p < 0.05), suggesting alterations in the glycolytic pathway which were confirmed by Western blot analyses of glycolytic enzymes. Infarct volume quantification showed a significant increase following N alone or N + OC as compared to saline control. Because glucose metabolism is critical for brain physiology, altered glycolysis deteriorates neural function, thus exacerbating ischemic brain damage.
Collapse
Affiliation(s)
- Francisca Diaz
- Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Ami P Raval
- Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA.,Peritz Scheinberg Cerebral Vascular Disease Research Laboratory, Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| |
Collapse
|
14
|
McCarthy M, Raval AP. The peri-menopause in a woman's life: a systemic inflammatory phase that enables later neurodegenerative disease. J Neuroinflammation 2020; 17:317. [PMID: 33097048 PMCID: PMC7585188 DOI: 10.1186/s12974-020-01998-9] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 10/14/2020] [Indexed: 02/08/2023] Open
Abstract
The peri-menopause or menopausal transition—the time period that surrounds the final years of a woman’s reproductive life—is associated with profound reproductive and hormonal changes in a woman’s body and exponentially increases a woman’s risk of cerebral ischemia and Alzheimer’s disease. Although our understanding of the exact timeline or definition of peri-menopause is limited, it is clear that there are two stages to the peri-menopause. These are the early menopausal transition, where menstrual cycles are mostly regular, with relatively few interruptions, and the late transition, where amenorrhea becomes more prolonged and lasts for at least 60 days, up to the final menstrual period. Emerging evidence is showing that peri-menopause is pro-inflammatory and disrupts estrogen-regulated neurological systems. Estrogen is a master regulator that functions through a network of estrogen receptors subtypes alpha (ER-α) and beta (ER-β). Estrogen receptor-beta has been shown to regulate a key component of the innate immune response known as the inflammasome, and it also is involved in regulation of neuronal mitochondrial function. This review will present an overview of the menopausal transition as an inflammatory event, with associated systemic and central nervous system inflammation, plus regulation of the innate immune response by ER-β-mediated mechanisms.
Collapse
Affiliation(s)
- Micheline McCarthy
- Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, 33136, USA
| | - Ami P Raval
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratory, Leonard M. Miller School of Medicine, University of Miami, 1420 NW 9th Avenue, Neurology Research Building, Room # 203H, Miami, FL, 33136, USA. .,Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, 33136, USA.
| |
Collapse
|
15
|
Azcoitia I, Barreto GE, Garcia-Segura LM. Molecular mechanisms and cellular events involved in the neuroprotective actions of estradiol. Analysis of sex differences. Front Neuroendocrinol 2019; 55:100787. [PMID: 31513774 DOI: 10.1016/j.yfrne.2019.100787] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 08/27/2019] [Accepted: 09/07/2019] [Indexed: 12/12/2022]
Abstract
Estradiol, either from peripheral or central origin, activates multiple molecular neuroprotective and neuroreparative responses that, being mediated by estrogen receptors or by estrogen receptor independent mechanisms, are initiated at the membrane, the cytoplasm or the cell nucleus of neural cells. Estrogen-dependent signaling regulates a variety of cellular events, such as intracellular Ca2+ levels, mitochondrial respiratory capacity, ATP production, mitochondrial membrane potential, autophagy and apoptosis. In turn, these molecular and cellular actions of estradiol are integrated by neurons and non-neuronal cells to generate different tissue protective responses, decreasing blood-brain barrier permeability, oxidative stress, neuroinflammation and excitotoxicity and promoting synaptic plasticity, axonal growth, neurogenesis, remyelination and neuroregeneration. Recent findings indicate that the neuroprotective and neuroreparative actions of estradiol are different in males and females and further research is necessary to fully elucidate the causes for this sex difference.
Collapse
Affiliation(s)
- Iñigo Azcoitia
- Department of Cell Biology, Faculty of Biology, Universidad Complutense de Madrid, 28040 Madrid, Spain; Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludables (CIBERFES), Instituto de Salud Carlos III, Avenida Monforte de Lemos, 3-5, 28029 Madrid, Spain.
| | - George E Barreto
- Department of Biological Sciences, School of Natural Sciences, University of Limerick, Limerick, Ireland.
| | - Luis M Garcia-Segura
- Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludables (CIBERFES), Instituto de Salud Carlos III, Avenida Monforte de Lemos, 3-5, 28029 Madrid, Spain; Instituto Cajal, CSIC, Avenida Doctor Arce 37, 28002 Madrid, Spain.
| |
Collapse
|
16
|
Saraf J, Sarmah D, Vats K, Kaur H, Pravalika K, Wanve M, Kalia K, Borah A, Dave KR, Yavagal DR, Bhattacharya P. Intra-arterial stem cell therapy modulates neuronal calcineurin and confers neuroprotection after ischemic stroke. Int J Neurosci 2019; 129:1039-1044. [PMID: 31203689 DOI: 10.1080/00207454.2019.1633315] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Aim: Calcineurin (CaN) is a threonine/phosphatase which play roles in neuronal homeostasis. Ischemic stroke induces hyperactivation of CaN which further triggers apoptotic signaling. CaN inhibition has limited therapeutic output and neurotoxicity due to its intricate roles in the neuronal network and requires a strategic modulation. Intra-arterial (IA) mesenchymal stem cells (MSCs) have shown to interact with the milieu in a paracrine manner as compared to CaN inhibitors to ameliorate the neuronal damage triggered by ischemia/reperfusion injury. The present study investigates the role of IA MSCs in modulating neuronal CaN after stroke onset. Materials and methods: To validate, middle-aged ovariectomized female rats exposed to MCAo (90 min) were treated with IA MSCs (1 × 105 MSCs) or phosphate-buffered saline (PBS) at 6 hours to check CaN expression in different groups.Tests for assessing functional and motor coordination were performed along with biochemical estimations. Furthermore, an inhibition study by non-selective inhibitor of neuronal calcium channel, flunarizine, was performed to explore the possible underlying mechanism by which IA MSCs may interact with CaN. Results: The study suggests that IA MSCs seemingly reduce the expression of CaN after ischemic stroke. IA MSCs have shown to improve the functional outcome and normalize oxidative parameters. Conclusion: Our study provides a preliminary evidence of role of IA MSCs in modulating CaN expression.
Collapse
Affiliation(s)
- Jackson Saraf
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad , Gandhinagar , Gujarat , India
| | - Deepaneeta Sarmah
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad , Gandhinagar , Gujarat , India
| | - Kanchan Vats
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad , Gandhinagar , Gujarat , India
| | - Harpreet Kaur
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad , Gandhinagar , Gujarat , India
| | - Kanta Pravalika
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad , Gandhinagar , Gujarat , India
| | - Madhuri Wanve
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad , Gandhinagar , Gujarat , India
| | - Kiran Kalia
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad , Gandhinagar , Gujarat , India
| | - Anupom Borah
- Cellular and Molecular Neurobiology Laboratory, Department of Life Science and Bioinformatics, Assam University , Silchar , Assam , India
| | - Kunjan R Dave
- Department of Neurology and Neurosurgery, University of Miami Miller School of Medicine , Miami , Florida , USA
| | - Dileep R Yavagal
- Department of Neurology and Neurosurgery, University of Miami Miller School of Medicine , Miami , Florida , USA
| | - Pallab Bhattacharya
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad , Gandhinagar , Gujarat , India
| |
Collapse
|
17
|
de Rivero Vaccari JP, Bramlett HM, Perez-Pinzon MA, Raval AP. Estrogen preconditioning: A promising strategy to reduce inflammation in the ischemic brain. CONDITIONING MEDICINE 2019; 2:106-113. [PMID: 32617523 PMCID: PMC7331970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
During the premenopausal phase of a woman's life, estrogen naturally protects against ischemic brain damage and its debilitating consequence of cognitive decline. However, the decline in estrogen at menopause exponentially increases a women's risk for cerebral ischemia and its severity. Supplementation of estrogen during menopause is the most logical solution to abate this increased risk for cerebral ischemia; however, continuous therapy has proven to be contraindicative. Studies from our laboratory over the past decade have shown that a single bolus or long-term periodic 17β-estradiol treatment(s) two days prior to ischemia mimics ischemic preconditioning-conferred protection of the brain in ovariectomized or reproductively senescent female rats. These studies also demonstrated that 17β-estradiol-induced preconditioning (EPC) requires estrogen receptor (ER)-subtype beta (ER-β) activation. ER-β is expressed throughout the brain, including in the hippocampus, which plays a key role in learning and memory. Because periodic activation of ER-β mitigates post-ischemic cognitive decline in ovariectomized female rats, it can be surmised that EPC has the potential to reduce post-ischemic damage and cognitive decline in females. Estrogens are key anti-inflammatory agents; therefore this review discusses the effects of EPC on the inflammasome. Furthermore, as we now clearly know, the brain acts differently in males and females. Indeed, neurodegenerative diseases, including cerebral ischemia, and pharmacological drugs affect males and females in different ways. Thus, inasmuch as the National Institutes of Health and the Stroke Treatment Academic Industry Roundtable (STAIR) consortium mandate inclusion of female experimental animals, this review also discusses the need to close the gap in our knowledge in future studies of EPC in female animal models of cerebral ischemia.
Collapse
Affiliation(s)
| | - Helen M. Bramlett
- Department of Neurological Surgery and The Miami Project to Cure Paralysis
- Bruce W. Carter Department of Veterans Affairs Medical Center, Miami
| | - Miguel A. Perez-Pinzon
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida 33136, U.S.A
| | - Ami P. Raval
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida 33136, U.S.A
| |
Collapse
|
18
|
Zafer D, Aycan N, Ozaydin B, Kemanli P, Ferrazzano P, Levine JE, Cengiz P. Sex differences in Hippocampal Memory and Learning following Neonatal Brain Injury: Is There a Role for Estrogen Receptor-α? Neuroendocrinology 2019; 109:249-256. [PMID: 30884486 PMCID: PMC6893032 DOI: 10.1159/000499661] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Accepted: 03/17/2019] [Indexed: 01/11/2023]
Abstract
Neonatal encephalopathy due to hypoxia-ischemia (HI) leads to severe, life-long morbidities in thousands of neonates born in the USA and worldwide each year. Varying capacities of long-term episodic memory, verbal working memory, and learning can present without cerebral palsy and have been associated with the severity of neonatal encephalopathy sustained at birth. Among children who sustain a moderate degree of HI at birth, girls have larger hippocampal volumes compared to boys. Clinical studies indicate that female neonatal brains are more resistant to the effects of neonatal HI, resulting in better long-term cognitive outcomes compared to males with comparable brain injury. Our most recent mechanistic studies have addressed the origins and cellular basis of sex differences in hippocampal neuroprotection following neonatal HI-related brain injury and implicate estrogen receptor-α (ERα) in the neurotrophin receptor-mediated hippocampal neuroprotection in female mice. This review summarizes the recent findings on ERα-dependent, neurotrophin-mediated hippocampal neuroprotection and weighs the evidence that this mechanism plays an important role in preservation of long-term memory and learning following HI in females.
Collapse
Affiliation(s)
- Dila Zafer
- Waisman Center, University of Wisconsin, Madison, Wisconsin, USA
| | - Nur Aycan
- Waisman Center, University of Wisconsin, Madison, Wisconsin, USA
| | - Burak Ozaydin
- Waisman Center, University of Wisconsin, Madison, Wisconsin, USA
| | - Pinar Kemanli
- Waisman Center, University of Wisconsin, Madison, Wisconsin, USA
| | - Peter Ferrazzano
- Waisman Center, University of Wisconsin, Madison, Wisconsin, USA
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, University of Wisconsin, Madison, Wisconsin, USA
| | - Jon E Levine
- Department of Neuroscience, University of Wisconsin, Madison, Wisconsin, USA
- Wisconsin National Primate Research Center, Madison, Wisconsin, USA
| | - Pelin Cengiz
- Waisman Center, University of Wisconsin, Madison, Wisconsin, USA,
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, University of Wisconsin, Madison, Wisconsin, USA,
| |
Collapse
|
19
|
Dave KR, Saul I, Raval AP, Perez-Pinzon MA. Preconditioning with CpG-ODN1826 reduces ischemic brain injury in young male mice: a replication study. CONDITIONING MEDICINE 2019; 2:178-184. [PMID: 32510041 PMCID: PMC7274220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Earlier studies established that ischemic tolerance can be induced in the brain using various strategies. An earlier study demonstrated that preconditioning with the toll-like receptor 9 ligand, CpG oligodeoxynucleotides (ODN), protects the brain against ischemic damage. To increase the potential translational value of the previous study, the goal of the present study was to replicate this earlier finding in a different animal cohort at a different site. In addition to these replication studies, following the Stroke Treatment Academic Industry Roundtable (STAIR) guidelines, we also conducted studies to evaluate the protective effect of CpG-ODN 1826 preconditioning on cerebral ischemic damage in ovariectomized (Ovx) female animals. Young male and female mice were treated with CpG-ODN 1826 or control ligand 3 days prior to the induction of transient (60 min) cerebral ischemia using a middle cerebral artery occlusion (MCAO) model. Infarct size was evaluated at ~24 h post-MCAO. We were able to replicate earlier findings that preconditioning with a low dose (20 μg/mouse) of CpG-ODN 1826 was able to lower cerebral ischemic damage in young male mice. However, we did not see any protective effect of low dose CpG-ODN 1826 preconditioning against cerebral ischemic damage in young Ovx female mice. Our study independently confirms the protective effect of CpG-ODN 1826 in inducing cerebral ischemia tolerance in male but not in Ovx female mice. Our study also demonstrates the feasibility of conducting such replication studies in rodent models of transient stroke.
Collapse
Affiliation(s)
- Kunjan R. Dave
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, University of Miami School of Medicine, Miami, FL, USA
- Department of Neurology, University of Miami School of Medicine, Miami, FL, USA
- Neuroscience Program, University of Miami School of Medicine, Miami, FL, USA
| | - Isabel Saul
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, University of Miami School of Medicine, Miami, FL, USA
- Department of Neurology, University of Miami School of Medicine, Miami, FL, USA
| | - Ami P. Raval
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, University of Miami School of Medicine, Miami, FL, USA
- Department of Neurology, University of Miami School of Medicine, Miami, FL, USA
- Neuroscience Program, University of Miami School of Medicine, Miami, FL, USA
| | - Miguel A. Perez-Pinzon
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, University of Miami School of Medicine, Miami, FL, USA
- Department of Neurology, University of Miami School of Medicine, Miami, FL, USA
- Neuroscience Program, University of Miami School of Medicine, Miami, FL, USA
| |
Collapse
|
20
|
Raval AP, Martinez CC, Mejias NH, de Rivero Vaccari JP. Sexual dimorphism in inflammasome-containing extracellular vesicles and the regulation of innate immunity in the brain of reproductive senescent females. Neurochem Int 2018; 127:29-37. [PMID: 30500463 DOI: 10.1016/j.neuint.2018.11.018] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 11/26/2018] [Accepted: 11/26/2018] [Indexed: 12/20/2022]
Abstract
A woman's risk for stroke increases exponentially following the onset of menopause; however, the underlying mechanisms responsible for the increased risk remain unknown. The depletion of endogenous estrogen at menopause is known to activate the inflammatory response. Therefore, in this study we have used reproductively senescent (RS) rats to test the hypotheses that (1) inflammasome activation is significantly higher in the brain of RS females (RSF) as compared to their younger counterparts and age-matched senescent male rats, and that (2) RS triggers an innate immune response mediated in part by inflammasome-containing extracellular vesicles (EV) that originate in the female reproductive organs and then spreads to the brain. We tested these hypotheses using male and female Sprague-Dawley rats (Young: 6-7 months and RS: 9-13 months). Hippocampus, gonads and serum were collected. Additionally, cerebrospinal fluid (CSF) of pre- and post-menopausal women (ages 23 to 37 and 52 to 68) was purchased and extracellular vesicles (EV) were isolated from serum and CSF. The Inflammasome proteins caspase-1, apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC) and IL-1β were then resolved by immunoblotting. We found that inflammasome protein expression increased significantly in the analyzed tissues in RSF as compared to young females (YF), such difference was not present in age-matched male rat brains. Interestingly, we found that Nik-related kinase (NRK), which is present in female reproductive organs was present in the CSF and serum-derived EV, suggesting that the source of the EV seen in the brain during RS/menopause originate, in part, in the female reproductive organs. Thus, this study shows for the first time an involvement of the inflammasome originating in the female reproductive system as a contributor to inflammation in the brain that makes the peri-menopausal women's brain more susceptible to neurodegenerative diseases such as stroke.
Collapse
Affiliation(s)
- Ami P Raval
- Cerebral Vascular Disease Research Laboratories, Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, 33136, USA.
| | - Camila C Martinez
- Department of Neurological Surgery and The Miami Project to Cure Paralysis, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, 33136, USA
| | - Nancy H Mejias
- Department of Neurological Surgery and The Miami Project to Cure Paralysis, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, 33136, USA
| | - Juan Pablo de Rivero Vaccari
- Department of Neurological Surgery and The Miami Project to Cure Paralysis, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, 33136, USA.
| |
Collapse
|
21
|
Whole Body Vibration Therapy after Ischemia Reduces Brain Damage in Reproductively Senescent Female Rats. Int J Mol Sci 2018; 19:ijms19092749. [PMID: 30217051 PMCID: PMC6164360 DOI: 10.3390/ijms19092749] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 09/03/2018] [Accepted: 09/05/2018] [Indexed: 12/14/2022] Open
Abstract
A risk of ischemic stroke increases exponentially after menopause. Even a mild-ischemic stroke can result in increased frailty. Frailty is a state of increased vulnerability to adverse outcomes, which subsequently increases risk of cerebrovascular events and severe cognitive decline, particularly after menopause. Several interventions to reduce frailty and subsequent risk of stroke and cognitive decline have been proposed in laboratory animals and patients. One of them is whole body vibration (WBV). WBV improves cerebral function and cognitive ability that deteriorates with increased frailty. The goal of the current study is to test the efficacy of WBV in reducing post-ischemic stroke frailty and brain damage in reproductively senescent female rats. Reproductively senescent Sprague-Dawley female rats were exposed to transient middle cerebral artery occlusion (tMCAO) and were randomly assigned to either WBV or no-WBV groups. Animals placed in the WBV group underwent 30 days of WBV (40 Hz) treatment performed twice daily for 15 min each session, 5 days each week. The motor functions of animals belonging to both groups were tested intermittently and at the end of the treatment period. Brains were then harvested for inflammatory markers and histopathological analysis. The results demonstrate a significant reduction in inflammatory markers and infarct volume with significant increases in brain-derived neurotrophic factor and improvement in functional activity after tMCAO in middle-aged female rats that were treated with WBV as compared to the no-WBV group. Our results may facilitate a faster translation of the WBV intervention for improved outcome after stroke, particularly among frail women.
Collapse
|
22
|
d'Adesky ND, de Rivero Vaccari JP, Bhattacharya P, Schatz M, Perez-Pinzon MA, Bramlett HM, Raval AP. Nicotine Alters Estrogen Receptor-Beta-Regulated Inflammasome Activity and Exacerbates Ischemic Brain Damage in Female Rats. Int J Mol Sci 2018; 19:ijms19051330. [PMID: 29710856 PMCID: PMC5983576 DOI: 10.3390/ijms19051330] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 04/23/2018] [Accepted: 04/24/2018] [Indexed: 12/14/2022] Open
Abstract
Smoking is a preventable risk factor for stroke and smoking-derived nicotine exacerbates post-ischemic damage via inhibition of estrogen receptor beta (ER-β) signaling in the brain of female rats. ER-β regulates inflammasome activation in the brain. Therefore, we hypothesized that chronic nicotine exposure activates the inflammasome in the brain, thus exacerbating ischemic brain damage in female rats. To test this hypothesis, adult female Sprague-Dawley rats (6–7 months old) were exposed to nicotine (4.5 mg/kg/day) or saline for 16 days. Subsequently, brain tissue was collected for immunoblot analysis. In addition, another set of rats underwent transient middle cerebral artery occlusion (tMCAO; 90 min) with or without nicotine exposure. One month after tMCAO, histopathological analysis revealed a significant increase in infarct volume in the nicotine-treated group (64.24 ± 7.3 mm3; mean ± SEM; n = 6) compared to the saline-treated group (37.12 ± 7.37 mm3; n = 7, p < 0.05). Immunoblot analysis indicated that nicotine increased cortical protein levels of caspase-1, apoptosis-associated speck-like protein containing a CARD (ASC) and pro-inflammatory cytokines interleukin (IL)-1β by 88% (p < 0.05), 48% (p < 0.05) and 149% (p < 0.05), respectively, when compared to the saline-treated group. Next, using an in vitro model of ischemia in organotypic slice cultures, we tested the hypothesis that inhibition of nicotine-induced inflammasome activation improves post-ischemic neuronal survival. Accordingly, slices were exposed to nicotine (100 ng/mL; 14–16 days) or saline, followed by treatment with the inflammasome inhibitor isoliquiritigenin (ILG; 24 h) prior to oxygen-glucose deprivation (OGD; 45 min). Quantification of neuronal death demonstrated that inflammasome inhibition significantly decreased nicotine-induced ischemic neuronal death. Overall, this study shows that chronic nicotine exposure exacerbates ischemic brain damage via activation of the inflammasome in the brain of female rats.
Collapse
Affiliation(s)
- Nathan D d'Adesky
- Cerebral Vascular Disease Research Center, Department of Neurology and Neuroscience Program (D4-5), P.O. Box 016960, University of Miami School of Medicine, Miami, FL 33101, USA.
| | - Juan Pablo de Rivero Vaccari
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami School of Medicine, Miami, FL 33136, USA.
| | - Pallab Bhattacharya
- Cerebral Vascular Disease Research Center, Department of Neurology and Neuroscience Program (D4-5), P.O. Box 016960, University of Miami School of Medicine, Miami, FL 33101, USA.
| | - Marc Schatz
- Cerebral Vascular Disease Research Center, Department of Neurology and Neuroscience Program (D4-5), P.O. Box 016960, University of Miami School of Medicine, Miami, FL 33101, USA.
| | - Miguel A Perez-Pinzon
- Cerebral Vascular Disease Research Center, Department of Neurology and Neuroscience Program (D4-5), P.O. Box 016960, University of Miami School of Medicine, Miami, FL 33101, USA.
| | - Helen M Bramlett
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami School of Medicine, Miami, FL 33136, USA.
- Bruce W. Carter Department of Veterans Affairs Medical Center, Miami, FL 33125, USA.
| | - Ami P Raval
- Cerebral Vascular Disease Research Center, Department of Neurology and Neuroscience Program (D4-5), P.O. Box 016960, University of Miami School of Medicine, Miami, FL 33101, USA.
| |
Collapse
|
23
|
Stradecki-Cohan HM, Cohan CH, Raval AP, Dave KR, Reginensi D, Gittens RA, Youbi M, Perez-Pinzon MA. Cognitive Deficits after Cerebral Ischemia and Underlying Dysfunctional Plasticity: Potential Targets for Recovery of Cognition. J Alzheimers Dis 2018; 60:S87-S105. [PMID: 28453486 DOI: 10.3233/jad-170057] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Cerebral ischemia affects millions of people worldwide and survivors suffer from long-term functional and cognitive deficits. While stroke and cardiac arrest are typically considered when discussing ischemic brain injuries, there is much evidence that smaller ischemic insults underlie neurodegenerative diseases, including Alzheimer's disease. The "regenerative" capacity of the brain relies on several aspects of plasticity that are crucial for normal functioning; less affected brain areas may take over function previously performed by irreversibly damaged tissue. To harness the endogenous plasticity mechanisms of the brain to provide recovery of cognitive function, we must first understand how these mechanisms are altered after damage, such as cerebral ischemia. In this review, we discuss the long-term cognitive changes that result after cerebral ischemia and how ischemia alters several plasticity processes. We conclude with a discussion of how current and prospective therapies may restore brain plasticity and allow for recovery of cognitive function, which may be applicable to several disorders that have a disruption of cognitive processing, including traumatic brain injury and Alzheimer's disease.
Collapse
Affiliation(s)
- Holly M Stradecki-Cohan
- Department of Neurology Cerebral Vascular Disease Research Laboratories, Miami, FL, USA.,Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Charles H Cohan
- Department of Neurology Cerebral Vascular Disease Research Laboratories, Miami, FL, USA
| | - Ami P Raval
- Department of Neurology Cerebral Vascular Disease Research Laboratories, Miami, FL, USA
| | - Kunjan R Dave
- Department of Neurology Cerebral Vascular Disease Research Laboratories, Miami, FL, USA.,Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Diego Reginensi
- Centro de Neurociencias, Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT AIP), City of Knowledge, Panama, Republic of Panama
| | - Rolando A Gittens
- Centro de Neurociencias, Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT AIP), City of Knowledge, Panama, Republic of Panama
| | - Mehdi Youbi
- Department of Neurology Cerebral Vascular Disease Research Laboratories, Miami, FL, USA
| | - Miguel A Perez-Pinzon
- Department of Neurology Cerebral Vascular Disease Research Laboratories, Miami, FL, USA.,Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
24
|
Cohan CH, Stradecki-Cohan HM, Morris-Blanco KC, Khoury N, Koronowski KB, Youbi M, Wright CB, Perez-Pinzon MA. Protein kinase C epsilon delays latency until anoxic depolarization through arc expression and GluR2 internalization. J Cereb Blood Flow Metab 2017; 37:3774-3788. [PMID: 28585865 PMCID: PMC5718329 DOI: 10.1177/0271678x17712178] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Global cerebral ischemia is a debilitating injury that damages the CA1 region of the hippocampus, an area important for learning and memory. Protein kinase C epsilon (PKCɛ) activation is a critical component of many neuroprotective treatments. The ability of PKCɛ activation to regulate AMPA receptors (AMPARs) remains unexplored despite the role of AMPARs in excitotoxicity after brain ischemia. We determined that PKCɛ activation increased expression of a protein linked to learning and memory, activity-regulated cytoskeleton-associated protein (arc). Also, arc is necessary for neuroprotection and confers protection through decreasing AMPAR currents via GluR2 internalization. In vivo, activation of PKCɛ increased arc expression through a BDNF/TrkB pathway, and decreased GluR2 mRNA levels. In hippocampal cultured slices, PKCɛ activation decreased AMPAR current amplitudes in an arc- and GluR2-dependent manner. Additionally, PKCɛ activation triggered an arc- and GluR2 internalization-dependent delay in latency until anoxic depolarization. Inhibiting arc also blocked PKCɛ-mediated neuroprotection against lethal oxygen and glucose deprivation. These data characterize a novel PKCɛ-dependent mechanism that for the first time defines a role for arc and AMPAR internalization in conferring neuroprotection.
Collapse
Affiliation(s)
- Charles H Cohan
- 1 Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA.,2 Evelyn F. McKnight Brain Institute, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA.,3 Department of Neurology, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA.,4 Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA
| | - Holly M Stradecki-Cohan
- 1 Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA.,3 Department of Neurology, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA.,4 Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA
| | - Kahlilia C Morris-Blanco
- 1 Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA.,3 Department of Neurology, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA.,4 Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA
| | - Nathalie Khoury
- 1 Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA.,3 Department of Neurology, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA.,4 Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA
| | - Kevin B Koronowski
- 1 Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA.,3 Department of Neurology, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA.,4 Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA
| | - Mehdi Youbi
- 1 Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA.,3 Department of Neurology, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA
| | - Clinton B Wright
- 2 Evelyn F. McKnight Brain Institute, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA.,3 Department of Neurology, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA.,4 Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA
| | - Miguel A Perez-Pinzon
- 1 Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA.,2 Evelyn F. McKnight Brain Institute, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA.,3 Department of Neurology, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA.,4 Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
25
|
Shi Z, Zhu L, Li T, Tang X, Xiang Y, Han X, Xia L, Zeng L, Nie J, Huang Y, Tsang CK, Wang Y, Lei Z, Xu Z, So KF, Ruan Y. Neuroprotective Mechanisms of Lycium barbarum Polysaccharides Against Ischemic Insults by Regulating NR2B and NR2A Containing NMDA Receptor Signaling Pathways. Front Cell Neurosci 2017; 11:288. [PMID: 29021742 PMCID: PMC5623723 DOI: 10.3389/fncel.2017.00288] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Accepted: 09/04/2017] [Indexed: 01/03/2023] Open
Abstract
Glutamate excitotoxicity plays an important role in neuronal death after ischemia. However, all clinical trials using glutamate receptor inhibitors have failed. This may be related to the evidence that activation of different subunit of NMDA receptor will induce different effects. Many studies have shown that activation of the intrasynaptic NR2A subunit will stimulate survival signaling pathways, whereas upregulation of extrasynaptic NR2B will trigger apoptotic pathways. A Lycium barbarum polysaccharide (LBP) is a mixed compound extracted from Lycium barbarum fruit. Recent studies have shown that LBP protects neurons against ischemic injury by anti-oxidative effects. Here we first reported that the effect of LBP against ischemic injury can be achieved by regulating NR2B and NR2A signaling pathways. By in vivo study, we found LBP substantially reduced CA1 neurons from death after transient global ischemia and ameliorated memory deficit in ischemic rats. By in vitro study, we further confirmed that LBP increased the viability of primary cultured cortical neurons when exposed to oxygen-glucose deprivation (OGD) for 4 h. Importantly, we found that LBP antagonized increase in expression of major proteins in the NR2B signal pathway including NR2B, nNOS, Bcl-2-associated death promoter (BAD), cytochrome C (cytC) and cleaved caspase-3, and also reduced ROS level, calcium influx and mitochondrial permeability after 4 h OGD. In addition, LBP prevented the downregulation in the expression of NR2A, pAkt and pCREB, which are important cell survival pathway components. Furthermore, LBP attenuated the effects of a NR2B co-agonist and NR2A inhibitor on cell mortality under OGD conditions. Taken together, our results demonstrated that LBP is neuroprotective against ischemic injury by its dual roles in activation of NR2A and inhibition of NR2B signaling pathways, which suggests that LBP may be a superior therapeutic candidate for targeting glutamate excitotoxicity for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Zhongshan Shi
- GHM Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China.,Department of Anatomy, Jinan University School of Medicine, Guangzhou, China.,Ministry of Education CNS Regeneration International Collaborative Laboratory, Jinan University, Guangzhou, China
| | - Lihui Zhu
- GHM Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China.,Department of Anatomy, Jinan University School of Medicine, Guangzhou, China.,Ministry of Education CNS Regeneration International Collaborative Laboratory, Jinan University, Guangzhou, China
| | - Tingting Li
- GHM Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, China
| | - Xiaoya Tang
- GHM Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, China
| | - Yonghui Xiang
- GHM Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, China
| | - Xinjia Han
- GHM Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, China
| | - Luoxing Xia
- GHM Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, China
| | - Ling Zeng
- GHM Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, China
| | - Junhua Nie
- GHM Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, China
| | - Yongxia Huang
- GHM Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, China.,Department of Ophthalmology and State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong, China
| | - Chi Kwan Tsang
- Clinical Neuroscience Institute, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Ying Wang
- Key Laboratory of South China Agricultural Plant Molecular Analysis and Genetic Improvement, Provincial Key Laboratory of Applied Botany, South China Botanical Garden, Chinese Academy of Sciences, Guangzhou, China
| | - Zhigang Lei
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Zaocheng Xu
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Kwok-Fai So
- GHM Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China.,Ministry of Education CNS Regeneration International Collaborative Laboratory, Jinan University, Guangzhou, China.,Department of Ophthalmology and State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong, China
| | - Yiwen Ruan
- GHM Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China.,Department of Anatomy, Jinan University School of Medicine, Guangzhou, China.,Ministry of Education CNS Regeneration International Collaborative Laboratory, Jinan University, Guangzhou, China
| |
Collapse
|
26
|
Li Z, Fang F, Wang Y, Wang L. Resveratrol protects CA1 neurons against focal cerebral ischemic reperfusion-induced damage via the ERK-CREB signaling pathway in rats. Pharmacol Biochem Behav 2016; 146-147:21-7. [PMID: 27143440 DOI: 10.1016/j.pbb.2016.04.007] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Revised: 04/06/2016] [Accepted: 04/29/2016] [Indexed: 12/22/2022]
Abstract
Ischemic stroke is a primary cause of mortality and disability in the aged population. Resveratrol (Res), a natural polyphenol enriched in plants, presents diverse biological activities, e.g., antiinflammatory and anti-oxidation effects. Here, we evaluated whether Res pretreatment influenced focal cerebral ischemia-induced cognitive impairment, and we explored the underlying mechanisms in rats. The results showed that a single administration of Res (30mg/kg, i.p.) at 1 or 4h, but not at 24h before focal cerebral ischemia exerted significant neuroprotective effects, including a reduction in hippocampal CA1 neuronal death and spatial cognition deficits caused by ischemia. The neuroprotective effects of Res were suppressed by pretreatment with MK801, an NMDA receptor blocker, or U0126, an extracellular signal regulated kinase 1/2 (ERK1/2) kinase inhibitor. A western blot analysis revealed that Res treatment at 1h before ischemia significantly increased ERK1/2 phosphorylation and cyclic-AMP response element binding protein (CREB) phosphorylation in the CA1 region of the hippocampus, which can be prevented with U0126 pretreatment. The results showed that the NMDA receptor-mediated ERK-CREB signaling pathway might participates in Res-induced neuroprotection in rats with focal cerebral ischemia.
Collapse
Affiliation(s)
- Zhen Li
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China; Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Fang Fang
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China; Pharmacy Intravenous Admiture Services, Binhu Hospital of Hefei, Anhui Medical University, Hefei, China
| | - Yuanyin Wang
- College of Stomatology, Anhui Medical University, Hefei, China
| | - Liecheng Wang
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China; Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China.
| |
Collapse
|
27
|
de Rivero Vaccari JP, Patel HH, Brand FJ, Perez-Pinzon MA, Bramlett HM, Raval AP. Estrogen receptor beta signaling alters cellular inflammasomes activity after global cerebral ischemia in reproductively senescence female rats. J Neurochem 2015; 136:492-6. [PMID: 26490364 DOI: 10.1111/jnc.13404] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2015] [Revised: 10/03/2015] [Accepted: 10/09/2015] [Indexed: 01/16/2023]
Abstract
Periodic treatments with estrogen receptor subtype-β (ER-β) agonist reduce post-ischemic hippocampal injury in ovariectomized rats. However, the underlying mechanism of how ER-β agonists protect the brain remains unknown. Global cerebral ischemia activates the innate immune response, and a key component of the innate immune response is the inflammasome. This study tests the hypothesis that ER-β regulates inflammasome activation in the hippocampus, thus reducing ischemic hippocampal damage in reproductively senescent female rats that received periodic ER-β agonist treatments. First, we determined the effect of hippocampal ER-β silencing on the expression of the inflammasome proteins caspase 1, apoptosis-associated speck-like protein containing a CARD (ASC), and interleukin (IL)-1β. Silencing of ER-β attenuated 17β-estradiol mediated decrease in caspase 1, ASC, and IL-1β. Next, we tested the hypothesis that periodic ER-β agonist treatment reduces inflammasome activation and ischemic damage in reproductively senescent female rats. Periodic ER-β agonist treatments significantly decreased inflammasome activation and increased post-ischemic live neuronal counts by 32% (p < 0.05) as compared to the vehicle-treated, reproductively senescent rats. Current findings demonstrated that ER-β activation regulates inflammasome activation and protects the brain from global ischemic damage in reproductively senescent female rats. Further investigation on the role of a periodic ER-β agonist regimen to reduce the innate immune response in the brain could help reduce the incidence and the impact of global cerebral ischemia in post-menopausal women. We propose that estrogen receptor subtype-β (ER-β) activation regulates inflammasome activation and protects the brain from global ischemic damage in reproductively senescent female rats.
Collapse
Affiliation(s)
- Juan Pablo de Rivero Vaccari
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami, Miami, Florida, USA
| | - Hersila H Patel
- Cerebral Vascular Disease Research Laboratories, Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Frank J Brand
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami, Miami, Florida, USA
| | - Miguel A Perez-Pinzon
- Cerebral Vascular Disease Research Laboratories, Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Helen M Bramlett
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami, Miami, Florida, USA.,Bruce W. Carter Department of Veterans Affairs Medical Center, University of Miami, Miami, Florida, USA
| | - Ami P Raval
- Cerebral Vascular Disease Research Laboratories, Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida, USA
| |
Collapse
|
28
|
Periodic Estrogen Receptor-Beta Activation: A Novel Approach to Prevent Ischemic Brain Damage. Neurochem Res 2014; 40:2009-17. [PMID: 24906488 DOI: 10.1007/s11064-014-1346-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Revised: 05/12/2014] [Accepted: 05/22/2014] [Indexed: 02/06/2023]
Abstract
In women, the risk for cerebral ischemia climbs rapidly after menopause. At menopause, production of ovarian hormones; i.e., progesterone and estrogen, slowly diminishes. Estrogen has been suggested to confer natural protection to premenopausal women from ischemic stroke and some of its debilitating consequences. This notion is also strongly supported by laboratory studies showing that a continuous chronic 17β-estradiol (E2; a potent estrogen) regimen protects brain from ischemic injury. However, concerns regarding the safety of the continuous intake of E2 were raised by the failed translation to the clinic. Recent studies demonstrated that repetitive periodic E2 pretreatments, in contrast to continuous E2 treatment, provided neuroprotection against cerebral ischemia in ovariectomized rats. Periodic E2 pretreatment protects hippocampal neurons through activation of estrogen receptor subtype beta (ER-β). Apart from neuroprotection, periodic activation of ER-β in ovariectomized rats significantly improves hippocampus-dependent learning and memory. Difficulties in learning and memory loss are the major consequence of ischemic brain damage. Periodic ER-β agonist pretreatment may provide pharmacological access to a protective state against ischemic stroke and its debilitating consequences. The use of ER-β-selective agonists constitutes a safer target for future research than ER-α agonist or E2, inasmuch as it lacks the ability to stimulate the proliferation of breast or endometrial tissue. In this review, we highlight ER-β signaling as a guide for future translational research to reduce cognitive decline and cerebral ischemia incidents/impact in post-menopausal women, while avoiding the side effects produced by chronic E2 treatment.
Collapse
|
29
|
Yavagal DR, Lin B, Raval AP, Garza PS, Dong C, Zhao W, Rangel EB, McNiece I, Rundek T, Sacco RL, Perez-Pinzon M, Hare JM. Efficacy and dose-dependent safety of intra-arterial delivery of mesenchymal stem cells in a rodent stroke model. PLoS One 2014; 9:e93735. [PMID: 24807059 PMCID: PMC4012944 DOI: 10.1371/journal.pone.0093735] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Accepted: 03/09/2014] [Indexed: 01/14/2023] Open
Abstract
Intra-arterial (IA) delivery of mesenchymal stem cells (MSCs) for acute ischemic stroke is attractive for clinical translation. However, studies using rat model of stroke have demonstrated that IA MSCs delivery can decrease middle cerebral artery (MCA) flow, which may limit its clinical translation. The goal of this study is to identify a dose of IA MSCs (maximum tolerated dose; MTD) that does not compromise MCA flow and evaluate its efficacy and optimal timing in a rat model of reversible middle cerebral artery occlusion (rMCAo). We sought to determine if there is a difference in efficacy of acute (1 h) versus sub-acute (24 h) IA MSCs treatment after rMCAo. Adult female Sprague-Dawley rats underwent rMCAo (90 min) and an hour later a single dose of MSCs (at de-escalating doses 1 × 10(6), 5 × 10(5), 2 × 10(5), 1 × 10(5) and 5 × 10(4)) was given using IA route. MSCs were suspended in phosphate buffered saline (PBS) and PBS alone was used for control experiments. We measured the percent change in mean laser Doppler flow signal over the ipsilateral MCA in de-escalating doses groups to determine MTD. The results demonstrated that the lowering of IA MSC dose to 1 × 10(5) and below did not compromise MCA flow and hence an IA MSC dose of 1 × 10(5) considered as MTD. Subsequently, 1 h and 24 h after rMCAo, rats were treated with IA MSCs or PBS. The 24 h delivery of IA MSCs significantly improved neurodeficit score and reduced the mean infarct volume at one month as compared to control, but not the 1 h delivery. Overall, this study suggests that the IA delivery of MSCs can be performed safely and efficaciously at the MTD of 1 × 10(5) delivered at 24 hours in rodent model of stroke.
Collapse
Affiliation(s)
- Dileep R. Yavagal
- Cerebral Vascular Disease Research Laboratories, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida, United States of America
- Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida, United States of America
- Interdisciplinary Stem Cell Institute, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida, United States of America
| | - Baowan Lin
- Cerebral Vascular Disease Research Laboratories, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida, United States of America
| | - Ami P. Raval
- Cerebral Vascular Disease Research Laboratories, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida, United States of America
- Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida, United States of America
| | - Philip S. Garza
- Cerebral Vascular Disease Research Laboratories, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida, United States of America
| | - Chuanhui Dong
- Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida, United States of America
| | - Weizhao Zhao
- Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida, United States of America
| | - Erika B. Rangel
- Interdisciplinary Stem Cell Institute, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida, United States of America
| | - Ian McNiece
- Interdisciplinary Stem Cell Institute, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida, United States of America
| | - Tatjana Rundek
- Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida, United States of America
| | - Ralph L. Sacco
- Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida, United States of America
| | - Miguel Perez-Pinzon
- Cerebral Vascular Disease Research Laboratories, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida, United States of America
- Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida, United States of America
| | - Joshua M. Hare
- Interdisciplinary Stem Cell Institute, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida, United States of America
- The Department of Medicine, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida, United States of America
| |
Collapse
|
30
|
Abstract
A transient, ischemia-resistant phenotype known as "ischemic tolerance" can be established in brain in a rapid or delayed fashion by a preceding noninjurious "preconditioning" stimulus. Initial preclinical studies of this phenomenon relied primarily on brief periods of ischemia or hypoxia as preconditioning stimuli, but it was later realized that many other stressors, including pharmacologic ones, are also effective. This review highlights the surprisingly wide variety of drugs now known to promote ischemic tolerance, documented and to some extent mechanistically characterized in preclinical animal models of stroke. Although considerably more experimentation is needed to thoroughly validate the ability of any currently identified preconditioning agent to protect ischemic brain, the fact that some of these drugs are already clinically approved for other indications implies that the growing enthusiasm for translational success in the field of pharmacologic preconditioning may be well justified.
Collapse
|
31
|
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
|
32
|
Raval AP, Borges-Garcia R, Javier Moreno W, Perez-Pinzon MA, Bramlett H. Periodic 17β-estradiol pretreatment protects rat brain from cerebral ischemic damage via estrogen receptor-β. PLoS One 2013; 8:e60716. [PMID: 23593292 PMCID: PMC3625208 DOI: 10.1371/journal.pone.0060716] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2012] [Accepted: 03/01/2013] [Indexed: 12/17/2022] Open
Abstract
Although chronic 17β-estradiol (E2) has been shown to be a cognition-preserving and neuroprotective agent in animal brain injury models, concern regarding its safety was raised by the failed translation of this phenomenon to the clinic. Previously, we demonstrated that a single bolus of E2 48 hr prior to ischemia protected the hippocampus from damage in ovariectomized rats via phosphorylation of cyclic-AMP response element binding protein, which requires activation of estrogen receptor subtype beta (ER-β). The current study tests the hypothesis that long-term periodic E2-treatment improves cognition and reduces post-ischemic hippocampal injury by means of ER-β activation. Ovariectomized rats were given ten injections of E2 at 48 hr intervals for 21 days. Hippocampal-dependent learning, memory and ischemic neuronal loss were monitored. Results demonstrated that periodic E2 treatments improved spatial learning, memory and ischemic neuronal survival in ovariectomized rats. Additionally, periodic ER-β agonist treatments every 48 hr improved post-ischemic cognition. Silencing of hippocampal ER-β attenuated E2-mediated ischemic protection suggesting that ER-β plays a key role in mediating the beneficial effects of periodic E2 treatments. This study emphasizes the need to investigate a periodic estrogen replacement regimen to reduce cognitive decline and cerebral ischemia incidents/impact in post-menopausal women.
Collapse
Affiliation(s)
- Ami P Raval
- Cerebral Vascular Disease Research Laboratories, Department of Neurology, University of Miami, Miami, Florida, USA.
| | | | | | | | | |
Collapse
|
33
|
Chen Y, Su Y, Run X, Sun Z, Wang T, Sun S, Liang Z. Pretreatment of PC12 Cells with 17β-estradiol Prevents Aβ-Induced Down-Regulation of CREB Phosphorylation and Prolongs Inhibition of GSK-3β. J Mol Neurosci 2012; 50:394-401. [DOI: 10.1007/s12031-012-9938-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2012] [Accepted: 12/05/2012] [Indexed: 10/27/2022]
|
34
|
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]
|
35
|
De Butte-Smith M, Zukin RS, Etgen AM. Effects of global ischemia and estradiol pretreatment on phosphorylation of Akt, CREB and STAT3 in hippocampal CA1 of young and middle-aged female rats. Brain Res 2012; 1471:118-28. [PMID: 22771860 DOI: 10.1016/j.brainres.2012.06.036] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2012] [Revised: 06/22/2012] [Accepted: 06/25/2012] [Indexed: 01/29/2023]
Abstract
Transient global ischemia induces selective, delayed neuronal death of pyramidal neurons in the hippocampal CA1. Whereas long term treatment of middle-aged female rats with estradiol at physiological doses ameliorates neuronal death, the signaling pathways that mediate the neuroprotection are, as yet, unknown. Protein kinase B (Akt) and downstream transcription factors, the cAMP response element binding protein (CREB) and signal transducer and activator of transcription (STAT3) are critical players in cellular survival following injury. The present study was undertaken to determine whether long term estradiol alters the phosphorylation status and activity of Akt, STAT3 and CREB in ovariohysterectomized, middle-aged and young female rats subjected to global ischemia. Irrespective of either hormone or ischemic condition, middle-aged females exhibited lower levels of p-CREB and higher levels of Akt and STAT3 in CA1 than young females, as assessed by Western blot. In middle-aged animals, ischemia increased the phosphorylation status/activity of Akt and STAT3, and decreased the phosphorylation status/activity of CREB in the hippocampal CA1. Whereas estradiol did not detectably alter the phosphorylation status/activity of Akt or STAT3, it prevented the ischemia-induced decrease in nuclear p-CREB. Similar results were observed for the young females. Collectively, these data demonstrate that CREB, STAT3, and Akt are involved in the molecular response to global ischemia and that age influences the status of CREB, STAT3 and Akt activity in CA1 under physiological as well as pathological conditions, further emphasizing the importance of including older rodents in neuroprotection studies.
Collapse
Affiliation(s)
- M De Butte-Smith
- Albert Einstein College of Medicine, Dominick P. Purpura Department of Neuroscience, Bronx, NY 10461, USA
| | | | | |
Collapse
|
36
|
Chronic nicotine exposure inhibits estrogen-mediated synaptic functions in hippocampus of female rats. Neurosci Lett 2012; 517:41-6. [PMID: 22521583 DOI: 10.1016/j.neulet.2012.04.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2011] [Revised: 03/24/2012] [Accepted: 04/05/2012] [Indexed: 11/24/2022]
Abstract
Nicotine, the addictive agent in cigarettes, reduces circulating estradiol-17β (E₂) and inhibits E₂-mediated intracellular signaling in hippocampus of female rats. In hippocampus, E₂-signaling regulates synaptic plasticity by phosphorylation of the N-methyl-D-aspartic acid receptor subunit NR2B and cyclic-AMP response element binding protein (pCREB). Therefore, we hypothesized that chronic nicotine exposure induces synaptic dysfunction in hippocampus of female rats. Female rats were exposed to nicotine or saline for 16 days followed by electrophysiological analysis of hippocampus. Briefly, population measurements of excitatory post-synaptic field potentials (fEPSPs) were recorded from stratum radiatum of the CA1 hippocampal slice subfield. A strict software-controlled protocol was used which recorded 30 min of baseline data (stimulation rate of 1/min), a paired-pulse stimulation sequence followed by tetanic stimulation, and 1h of post-tetanus recording. EPSP amplitude and the initial EPSP slope were measured off-line. We then investigated by Western blot analysis the effects of nicotine on hippocampal estrogen receptor-beta (ER-β), NR2B and pCREB. The results demonstrated significantly decreased post-tetanic potentiation and paired-pulse facilitation at the 40, and 80 ms interval in nicotine-exposed rats compared to the saline group. Western blot analysis revealed that nicotine decreased protein levels of ER-β, NR2B, and pCREB. We also confirmed the role of E₂ in regulating NR2B and pCREB phosphorylation by performing Western blots in hippocapmal tissue obtained from E₂-treated ovariectomized rats. In conclusion, chronic nicotine exposure attenuates short-term synaptic plasticity, and the observed synaptic defects might be a consequence of loss of estradiol-17β-signaling. However, determining the exact molecular mechanisms of chronic nicotine exposure on synaptic plasticity specific to the female brain require further investigation.
Collapse
|
37
|
Shi GX, Andres DA, Cai W. Ras family small GTPase-mediated neuroprotective signaling in stroke. Cent Nerv Syst Agents Med Chem 2012; 11:114-37. [PMID: 21521171 DOI: 10.2174/187152411796011349] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2010] [Revised: 01/18/2011] [Accepted: 03/22/2011] [Indexed: 12/31/2022]
Abstract
Selective neuronal cell death is one of the major causes of neuronal damage following stroke, and cerebral cells naturally mobilize diverse survival signaling pathways to protect against ischemia. Importantly, therapeutic strategies designed to improve endogenous anti-apoptotic signaling appear to hold great promise in stroke treatment. While a variety of complex mechanisms have been implicated in the pathogenesis of stroke, the overall mechanisms governing the balance between cell survival and death are not well-defined. Ras family small GTPases are activated following ischemic insults, and in turn, serve as intrinsic switches to regulate neuronal survival and regeneration. Their ability to integrate diverse intracellular signal transduction pathways makes them critical regulators and potential therapeutic targets for neuronal recovery after stroke. This article highlights the contribution of Ras family GTPases to neuroprotective signaling cascades, including mitogen-activated protein kinase (MAPK) family protein kinase- and AKT/PKB-dependent signaling pathways as well as the regulation of cAMP response element binding (CREB), Forkhead box O (FoxO) and hypoxiainducible factor 1(HIF1) transcription factors, in stroke.
Collapse
Affiliation(s)
- Geng-Xian Shi
- Department of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, 741 S. Limestone St., Lexington, KY 40536-0509, USA.
| | | | | |
Collapse
|
38
|
Raval AP, Dave KR, Saul I, Gonzalez GJ, Diaz F. Synergistic inhibitory effect of nicotine plus oral contraceptive on mitochondrial complex-IV is mediated by estrogen receptor-β in female rats. J Neurochem 2012; 121:157-67. [DOI: 10.1111/j.1471-4159.2012.07661.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
39
|
Johnsen D, Murphy SJ. Isoflurane preconditioning protects neurons from male and female mice against oxygen and glucose deprivation and is modulated by estradiol only in neurons from female mice. Neuroscience 2011; 199:368-74. [PMID: 21985935 DOI: 10.1016/j.neuroscience.2011.09.053] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2011] [Revised: 09/22/2011] [Accepted: 09/25/2011] [Indexed: 11/19/2022]
Abstract
The volatile anesthetic, isoflurane, can protect the brain if administered before an insult such as an ischemic stroke. However, this protective "preconditioning" response to isoflurane is specific to males, with females showing an increase in brain damage following isoflurane preconditioning and subsequent focal cerebral ischemia. Innate cell sex is emerging as an important player in neuronal cell death, but its role in the sexually dimorphic response to isoflurane preconditioning has not been investigated. We used an in vitro model of isoflurane preconditioning and ischemia (oxygen and glucose deprivation, OGD) to test the hypotheses that innate cell sex dictates the response to isoflurane preconditioning and that 17β-estradiol attenuates any protective effect from isoflurane preconditioning in neurons via nuclear estrogen receptors. Sex-segregated neuron cultures derived from postnatal day 0-1 mice were exposed to either 0% or 3% isoflurane preconditioning for 1 h. In separate experiments, 17β-estradiol and the non-selective estrogen receptor antagonist ICI 182,780 were added 24 h before preconditioning and then removed at the end of the preconditioning period. Twenty-three hours after preconditioning, all cultures underwent 2 h of OGD. Twenty-four hours following OGD, cell viability was quantified using calcein-AM fluorescence. We observed that isoflurane preconditioning increased cell survival following subsequent OGD regardless of innate cell sex, but that the presence of 17β-estradiol before and during isoflurane preconditioning attenuated this protection only in female neurons independent of nuclear estrogen receptors. We also found that independent of preconditioning treatment, female neurons were less sensitive to OGD compared with male neurons and that transient treatment with 17β-estradiol protected both male and female neurons from subsequent OGD. More studies are needed to determine how cell type, cell sex, and sex steroids like 17β-estradiol may impact on anesthetic preconditioning and subsequent ischemic outcomes in the brain.
Collapse
Affiliation(s)
- D Johnsen
- Department of Anesthesiology and Perioperative Medicine, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Mail Code: UHN-2, Portland, OR 97239, USA
| | | |
Collapse
|
40
|
Etgen AM, Jover-Mengual T, Zukin RS. Neuroprotective actions of estradiol and novel estrogen analogs in ischemia: translational implications. Front Neuroendocrinol 2011; 32:336-52. [PMID: 21163293 PMCID: PMC3080451 DOI: 10.1016/j.yfrne.2010.12.005] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2010] [Revised: 12/06/2010] [Accepted: 12/09/2010] [Indexed: 11/15/2022]
Abstract
This review highlights our investigations into the neuroprotective efficacy of estradiol and other estrogenic agents in a clinically relevant animal model of transient global ischemia, which causes selective, delayed death of hippocampal CA1 neurons and associated cognitive deficits. We find that estradiol rescues a significant number of CA1 pyramidal neurons that would otherwise die in response to global ischemia, and this is true when hormone is provided as a long-term pretreatment at physiological doses or as an acute treatment at the time of reperfusion. In addition to enhancing neuronal survival, both forms of estradiol treatment induce measurable cognitive benefit in young animals. Moreover, estradiol and estrogen analogs that do not bind classical nuclear estrogen receptors retain their neuroprotective efficacy in middle-aged females deprived of ovarian hormones for a prolonged duration (8weeks). Thus, non-feminizing estrogens may represent a new therapeutic approach for treating the neuronal damage associated with global ischemia.
Collapse
Affiliation(s)
- Anne M Etgen
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA.
| | | | | |
Collapse
|
41
|
|
42
|
Ji Y, Tang B, Traub RJ. Spinal estrogen receptor alpha mediates estradiol-induced pronociception in a visceral pain model in the rat. Pain 2011; 152:1182-1191. [PMID: 21392887 DOI: 10.1016/j.pain.2011.01.046] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2010] [Revised: 01/21/2011] [Accepted: 01/24/2011] [Indexed: 02/07/2023]
Abstract
We previously reported that 17β-estradiol (E2) is pronociceptive in a visceral pain model in the rat. Subcutaneously (s.c.) administered E2 reversed the decrease in the colorectal distention (CRD)-evoked visceromotor response produced by ovariectomy (OVx) and CRD-induced nociceptive responses were greater in proestrous rats compared with met/diestrous rats. The site of action, the type of estrogen receptors activated, and the possible intracellular signaling pathway involved are yet to be established. In the present study, intrathecal (i.t.) E2 administered to OVx rats mimicked the effects of s.c. E2, suggesting that spinal estrogen receptors are involved. This is further supported by the observations that the anti-estrogen ICI 182,780 injected i.t. in intact female rats significantly decreased the visceromotor response to CRD, the response of colonic afferents was not affected by OVx, and colonic afferents did not label for estrogen receptor α (ERα). The ERα selective agonist, 4,4',4''-[4-propyl-(1H)-pyrazole-1,3,5-triyl]tris-phenol (PPT; s.c. or i.t.) facilitated the visceromotor response similar to E2, suggesting ERα activation is involved in mediating the pronociceptive effect of E2. PPT (s.c. or i.t.) increased the response of spinal dorsal horn neurons to CRD, indicating a spinal site of action. In addition, s.c. E2 or PPT increased CRD-induced spinal extracellular signal-regulated kinase (ERK) phosphorylation that was not observed in OVx rats and a mitogen-activated protein kinase (MAPK) kinase (MEK) inhibitor blocked facilitation of the visceromotor response by PPT. Taken together, the present study demonstrates that spinal ERα mediates the pronociceptive effect of E2 on visceral signal processing through activation of the MAPK pathway.
Collapse
Affiliation(s)
- Yaping Ji
- Department of Neural and Pain Sciences, University of Maryland Dental School, Baltimore, MD, USA
| | | | | |
Collapse
|
43
|
Raval AP, Hirsch N, Dave KR, Yavagal DR, Bramlett H, Saul I. Nicotine and estrogen synergistically exacerbate cerebral ischemic injury. Neuroscience 2011; 181:216-25. [PMID: 21334425 DOI: 10.1016/j.neuroscience.2011.02.036] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2010] [Revised: 01/16/2011] [Accepted: 02/15/2011] [Indexed: 02/07/2023]
Abstract
The greater incidence of myocardial infarction, cardiac arrest, and ischemic stroke among women who smoke and use oral contraception (OC) compared to women who do not smoke and who do or do not use OC may be due in part to how nicotine influences endocrine function in women. For example, we recently demonstrated that chronic exposure to nicotine, the addictive agent in tobacco smoke responsible for the elevated risk of cardiac arrest, abolishes the endogenous or exogenous 17β-estradiol-conferred protection of the hippocampus against global cerebral ischemia (a potential outcome of cardiac arrest) in naive or ovariectomized female rats. In the current study we examined the hypotheses that (1) a synergistic deleterious effect of nicotine plus oral contraceptives exacerbates post-ischemic hippocampal damage in female rats, and (2) nicotine directly inhibits estrogen-mediated intracellular signaling in the hippocampus. To test first hypothesis and to simulate smoking behavior-induced nicotine levels in the human body, we implanted osmotic pumps containing nicotine in the female rats for 16 days. Furthermore, we mimicked the use of oral contraceptives in females by administering oral contraceptives orally to the rat. Rats exposed to either nicotine alone or in combination with oral contraceptives were subjected to an episode of cerebral ischemia and the resultant brain damage was quantified. These results showed for the first time that nicotine with oral contraceptives did indeed exacerbate post-ischemic CA1 damage as compared to nicotine alone in naive female rats. In ex vivo hippocampal slice cultures, we found that nicotine alone or with 17β-estradiol directly hinders estrogen receptors-mediated phosphorylation of cyclic-AMP element binding protein, a process required for neuronal survival and also exacerbates ischemic damage. Thus, nicotine can affect the outcome of cerebral ischemia by influencing brain endocrine function directly rather than through indirect systemic effects.
Collapse
Affiliation(s)
- A P Raval
- Cerebral Vascular Disease Research Laboratories, Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL-33136, USA.
| | | | | | | | | | | |
Collapse
|
44
|
Bakkum BW, Fan L, Pandey SC, Cohen RS. Hetereogeneity of dose and time effects of estrogen on neuron-specific neuronal protein and phosphorylated cyclic AMP response element-binding protein in the hippocampus of ovariectomized rats. J Neurosci Res 2011; 89:883-97. [PMID: 21337376 DOI: 10.1002/jnr.22601] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2010] [Revised: 12/20/2010] [Accepted: 12/21/2010] [Indexed: 12/23/2022]
Abstract
Previous studies have shown changes in the cyclic AMP response element-binding protein (CREB) signaling pathway in CA1 and CA3 regions of the rostral hippocampus with 10 μg estrogen treatment for 14 days. It appears that estrogen's action on CREB phosphorylation in brain structures depends on other estrogen doses and lengths of treatment. We therefore examined the effects of moderate regimens [2.5 μg estradiol benzoate (EB) for 4 or 14 days] on mean numbers of neuron-specific neuronal protein (NeuN)-positive cells and phosphorylated CREB (pCREB)-positive cells and subregion volume defined by NeuN and pCREB immunolabeling and compared those results with results from the high regimen (10 μg EB for 14 days) in CA1, CA2, and CA3 regions and dorsal (DDG) and ventral (VDG) dentate gyrus and hilus of the hippocampus of ovariectomized rats by stereology. For whole hippocampus, all regimens increased mean neuronal (NeuN) numbers and pCREB-positive cell and volume compared with sesame oil (SO) in CA1, CA2, and CA3 regions, DDG and VDG, and hilus. In rostral hippocampus, however, some hippocampal subregions were not responsive to the high regimen, and the moderate regimens appear to be more effective for increasing mean number of NeuN-positive neurons and pCREB-positive cells and subregion volume. Heterogeneity in responsiveness to estrogen was mainly seen within rostral, but not whole, hippocampal subregions. Our results indicate that responsiveness of cells expressing NeuN and pCREB to different EB regimens may vary depending on the specific region of the hippocampus.
Collapse
Affiliation(s)
- Barclay W Bakkum
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, Illinois; Illinois College of Optometry, Chicago, IL 60612, USA
| | | | | | | |
Collapse
|
45
|
Hippocampal neuronal nitric oxide synthase (nNOS) is regulated by nicotine and stress in female but not in male rats. Brain Res 2011; 1368:134-42. [DOI: 10.1016/j.brainres.2010.10.090] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2010] [Revised: 10/21/2010] [Accepted: 10/26/2010] [Indexed: 12/14/2022]
|
46
|
Yang LC, Zhang QG, Zhou CF, Yang F, Zhang YD, Wang RM, Brann DW. Extranuclear estrogen receptors mediate the neuroprotective effects of estrogen in the rat hippocampus. PLoS One 2010; 5:e9851. [PMID: 20479872 PMCID: PMC2866326 DOI: 10.1371/journal.pone.0009851] [Citation(s) in RCA: 143] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2010] [Accepted: 03/04/2010] [Indexed: 11/26/2022] Open
Abstract
Background 17β-estradiol (E2) has been implicated to exert neuroprotective effects in the brain following cerebral ischemia. Classically, E2 is thought to exert its effects via genomic signaling mediated by interaction with nuclear estrogen receptors. However, the role and contribution of extranuclear estrogen receptors (ER) is unclear and was the subject of the current study. Methodology/Principal Findings To accomplish this goal, we employed two E2 conjugates (E2 dendrimer, EDC, and E2-BSA) that can interact with extranuclear ER and exert rapid nongenomic signaling, but lack the ability to interact with nuclear ER due to their inability to enter the nucleus. EDC or E2-BSA (10 µM) was injected icv 60 min prior to global cerebral ischemia (GCI). FITC-tagged EDC or E2-BSA revealed high uptake in the hippocampal CA1 region after icv injection, with a membrane (extranuclear) localization pattern in cells. Both EDC and E2-BSA exerted robust neuroprotection in the CA1 against GCI, and the effect was blocked by the ER antagonist, ICI182,780. EDC and E2-BSA both rapidly enhanced activation of the prosurvival kinases, ERK and Akt, while attenuating activation of the proapoptotic kinase, JNK following GCI, effects that were blocked by ICI182,780. Administration of an MEK or PI3K inhibitor blocked the neuroprotective effects of EDC and E2-BSA. Further studies showed that EDC increased p-CREB and BDNF in the CA1 region in an ERK- and Akt-dependent manner, and that cognitive outcome after GCI was preserved by EDC in an ER-dependent manner. Conclusions/Significance In conclusion, the current study demonstrates that activation of extranuclear ER results in induction of ERK-Akt-CREB-BDNF signaling in the hippocampal CA1 region, which significantly reduces ischemic neuronal injury and preserves cognitive function following GCI. The study adds to a growing literature that suggests that extranuclear ER can have important actions in the brain.
Collapse
Affiliation(s)
- Li-cai Yang
- Experimental and Research Center, North China Coal Medical University, Tangshan, Hebei, People's Republic of China
| | - Quan-Guang Zhang
- Institute of Molecular Medicine and Genetics, Medical College of Georgia, Augusta, Georgia, United States of America
| | - Cai-feng Zhou
- Experimental and Research Center, North China Coal Medical University, Tangshan, Hebei, People's Republic of China
| | - Fang Yang
- Experimental and Research Center, North China Coal Medical University, Tangshan, Hebei, People's Republic of China
| | - Yi-dong Zhang
- Experimental and Research Center, North China Coal Medical University, Tangshan, Hebei, People's Republic of China
| | - Rui-min Wang
- Experimental and Research Center, North China Coal Medical University, Tangshan, Hebei, People's Republic of China
- * E-mail: (RMW); (DWB)
| | - Darrell W. Brann
- Institute of Molecular Medicine and Genetics, Medical College of Georgia, Augusta, Georgia, United States of America
- * E-mail: (RMW); (DWB)
| |
Collapse
|
47
|
Jover-Mengual T, Miyawaki T, Latuszek A, Alborch E, Zukin RS, Etgen AM. Acute estradiol protects CA1 neurons from ischemia-induced apoptotic cell death via the PI3K/Akt pathway. Brain Res 2010; 1321:1-12. [PMID: 20114038 DOI: 10.1016/j.brainres.2010.01.046] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2009] [Revised: 12/03/2009] [Accepted: 01/18/2010] [Indexed: 11/28/2022]
Abstract
Global ischemia arising during cardiac arrest or cardiac surgery causes highly selective, delayed death of hippocampal CA1 neurons. Exogenous estradiol ameliorates global ischemia-induced neuronal death and cognitive impairment in male and female rodents. However, the molecular mechanisms by which a single acute injection of estradiol administered after the ischemic event intervenes in global ischemia-induced apoptotic cell death are unclear. Here we show that acute estradiol acts via the phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) signaling cascade to protect CA1 neurons in ovariectomized female rats. We demonstrate that global ischemia promotes early activation of glycogen synthase kinase-3beta (GSK3beta) and forkhead transcription factor of the O class (FOXO)3A, known Akt targets that are related to cell survival, and activation of caspase-3. Estradiol prevents ischemia-induced dephosphorylation and activation of GSK3beta and FOXO3A, and the caspase death cascade. These findings support a model whereby estradiol acts by activation of PI3K/Akt signaling to promote neuronal survival in the face of global ischemia.
Collapse
Affiliation(s)
- Teresa Jover-Mengual
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA.
| | | | | | | | | | | |
Collapse
|
48
|
Raval AP, Bhatt A, Saul I. Chronic nicotine exposure inhibits 17beta-estradiol-mediated protection of the hippocampal CA1 region against cerebral ischemia in female rats. Neurosci Lett 2009; 458:65-9. [PMID: 19442878 DOI: 10.1016/j.neulet.2009.04.021] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2009] [Revised: 04/09/2009] [Accepted: 04/09/2009] [Indexed: 10/20/2022]
Abstract
Nicotine addiction in women increases the risk of ischemic stroke. Importantly, women who smoke and use hormone replacement therapy/oral contraceptives greatly increase their risk of coronary heart disease and ischemic stroke as compared to nonsmoking women who use occasionally oral contraceptives. Nicotine addiction disturbs the normal periodicity of the menstrual cycle and induces early onset of menopause in women; however, the mechanism of the synergistic effects of nicotine and sex hormones on cerebrovascular health is not clearly understood. In the current study based on a rat model of global cerebral ischemia, our goals are (1) to determine whether chronic nicotine exposure abrogates beneficial effects of estrogen on hippocampal neurons subjected to ischemia, and (2) to determine whether nicotine exposure antagonizes estrogen signaling by reducing the availability of estrogen receptor(s). To test the effects of chronic nicotine exposure, normally cycling or ovariectomized rats were injected with nicotine daily for 15 days. To investigate the efficacy of estrogen treatment, nicotine-exposed ovariectomized rats were injected with a bolus of 17beta-estradiol and 48h later ischemia was induced. Our results demonstrated that chronic nicotine exposure followed by ischemic insult at the proestrus stage of the estrous cycle showed that only 14% of normal neurons remained compared to the non-nicotine-treated group (p<0.05). Similarly, a bolus of 17beta-estradiol to nicotine-treated ovariectomized rats showed only 26% of normal neurons remaining as against 47% in the non-nicotine-treated group. Nicotine exposure decreased ERbeta but not ERalpha protein levels in the hippocampus, suggesting a role for ERbeta in increased post-ischemic neurodegeneration from nicotine exposure.
Collapse
Affiliation(s)
- Ami P Raval
- Cerebral Vascular Disease Research Center, Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33101, USA.
| | | | | |
Collapse
|