1
|
Foster TC, Kumar A. Sex, senescence, senolytics, and cognition. Front Aging Neurosci 2025; 17:1555872. [PMID: 40103928 PMCID: PMC11913825 DOI: 10.3389/fnagi.2025.1555872] [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: 01/05/2025] [Accepted: 02/11/2025] [Indexed: 03/20/2025] Open
Abstract
This review focuses on sexual dimorphism in cellular senescence and senolytic treatment in relation to brain health and age-related cognitive decline. The stressors of aging, DNA damage, inflammation, and oxidative stress induce cell senescence, a hallmark of aging. Senescent cells change their function and molecular profile and are primed to release pro-inflammatory cytokines. The functional changes include the activation of cell signals to prevent cell death. The release of pro-inflammatory cytokines from peripheral senescent cells during middle age induces senescence of neighbor cells and heightens the level of systemic inflammation, contributing to neuroinflammation. In response to neuroinflammation and oxidative stress, some neurons alter their physiology, decreasing neuronal excitability and synaptic transmission. Senescent neurophysiology is protective against cell death due to excitotoxicity, at the expense of a loss of normal cell function, contributing to age-related cognitive decline. The level of peripheral cell senescence and systemic inflammation may underlie sexual dimorphism in the prevalence, symptoms, and pathogenesis of age-related diseases, including neurodegenerative diseases. Sex differences have been observed for senescence of astrocytes, microglia, and peripheral cells, including those involved in innate and adaptive immune responses. Interventions that remove senescent cells, such as senolytic drugs, can reduce or ameliorate some of the aging-related loss of function. Similarities and differences in senolytic responses of males and females depend on the system examined, the treatment regimen, the level of senescent cell burden, and the age when treatment is initiated. Estrogen impacts several of these factors and influences the transcription of genes promoting growth, proliferation, and cell survival programs in a manner opposite that of senolytic drugs. In addition, estrogen has anti-aging effects that are independent of cell senescence, including rapidly modifying senescent neurophysiology. Thus, it is important to recognize that, in addition to sex differences in cell senescence, there are other sexually dimorphic mechanisms that contribute to the aging process. The results indicate that senolytics interact with fundamental biology, including sex hormones.
Collapse
Affiliation(s)
- Thomas C Foster
- McKnight Brain Institute, Department of Neuroscience, University of Florida, Gainesville, FL, United States
- Genetics and Genomics Graduate Program, Genetics Institute, University of Florida, Gainesville, FL, United States
| | - Ashok Kumar
- McKnight Brain Institute, Department of Neuroscience, University of Florida, Gainesville, FL, United States
| |
Collapse
|
2
|
Szychowski KA, Skóra B. Disruption of neurosteroid synthesis and release by tris(2,3-dibromopropyl)isocyanurate in primary mouse cortical astrocytes in vitro. J Appl Toxicol 2023; 43:1604-1612. [PMID: 37254606 DOI: 10.1002/jat.4498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/08/2023] [Accepted: 05/15/2023] [Indexed: 06/01/2023]
Abstract
Neurosteroidogenesis in astrocytes is crucial for the proper development and functioning of the brain. During this process, key neurohormones such as progesterone (P4 ), testosterone (T), and estradiol (E2 ) are produced. Proper production and release of neurosteroids can be affected by substances referred to as endocrine-disrupting compounds (EDCs). Tris-(2,3-dibromopropyl)isocyanurate (TBC) is a representative of novel brominated flame retardants used to stop ignition or reduce fire-related property damage to plastics, polyolefin, polyphenyl alkene, unsaturated polyester, synthetic rubber, and fibers. Interestingly, previous studies have shown that TBC can enhance the proliferation of estradiol-sensitive breast cancers in vitro, which suggests that TBC has EDC properties. Therefore, given the suspected endocrine-disrupting properties of TBC, the aim of the present study was to determine the impact of TBC on the neurosteroid (P4 , T, and E2 ) production and secretion as well as the mRNA expression of key enzymes involved in its production in mouse astrocytes in vitro. Our paper shows that TBC increases P4 production with a strong decrease in T production, which is accompanied by a decrease in Cyp17a1 mRNA expression, that is, the main enzyme metabolizing P4 to T. Moreover, TBC in both studied concentrations increases P4 secretion in the culture medium. Finally, our studies have demonstrated an increase in the expression of Cyp19a1 mRNA, an enzyme metabolizing T to E2 , with a simultaneous increase in the amount of E2 in cells. Our data clearly show that TBC in an in vitro environment acts as EDCs, which may lead to serious consequences for the proper development and functioning of the brain.
Collapse
Affiliation(s)
- Konrad A Szychowski
- Department of Biotechnology and Cell Biology, Medical College, University of Information Technology and Management in Rzeszow, Rzeszow, Poland
| | - Bartosz Skóra
- Department of Biotechnology and Cell Biology, Medical College, University of Information Technology and Management in Rzeszow, Rzeszow, Poland
| |
Collapse
|
3
|
Snyder K, Gorse K, Kochanek PM, Jackson TC. Neuronal RBM5 modulates cell signaling responses to traumatic and hypoxic-ischemic injury in a sex-dependent manner. Cell Death Discov 2023; 9:379. [PMID: 37848418 PMCID: PMC10582027 DOI: 10.1038/s41420-023-01677-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 09/22/2023] [Accepted: 10/03/2023] [Indexed: 10/19/2023] Open
Abstract
It is not clear if inhibiting the pro-death gene RNA binding motif 5 (RBM5) is neuroprotective in isolated primary neurons or if it regulates cell survival in a sex-dependent manner. Here we established sex-dichotomized primary cortical neuron cultures from transgenic mice harboring a floxed RBM5 gene-trap. Lentivirus-mediated expression of CRE was used to silence RBM5 expression. Male and female neurons were maintained in next-generation Neurobasal-Plus media and subjected to a mechanical stretch-injury (to model traumatic brain injury) or oxygen-glucose deprivation/OGD (to model ischemia). RBM5 KO did not affect 24 h post-injury survival as determined by lactate dehydrogenase (LDH) release, in either paradigm. In contrast, female KO neurons had increased spectrin breakdown products post-insult (in both models). Furthermore, in OGD, RBM5 KO in male neurons exacerbated injury-induced downregulation of pro-survival AKT activation (pAKT473) but conversely led to pAKT473 sparing in female neurons. Moreover, global proteomics identified 19 differentially expressed (DE) proteins in OGD-injured male neurons, and 102 DE proteins in injured female neurons. Two novel RBM5-regulated proteins (PIGQ and EST1C) were identified in injured male KO neurons, and 8 novel proteins identified in injured female KO neurons (S35A5, DHTK1, STX3, IF3M, RN167, K1C14, DYHS, and MED13). In summary, RBM5 inhibition does not modify neuronal survival in primary mouse neurons in 2 clinically relevant models of excitotoxic insult, but RBM5 does regulate intracellular responses to injury in a sex-dependent manner.
Collapse
Affiliation(s)
- Kara Snyder
- University of South Florida, Morsani College of Medicine, USF Health Heart Institute, MDD 0630, 560 Channelside Dr, Tampa, FL, 33602, USA
- University of South Florida, Morsani College of Medicine, Department of Molecular Pharmacology & Physiology, 12901 Bruce B Downs Blvd, Tampa, FL, 33612, USA
| | - Kiersten Gorse
- University of South Florida, Morsani College of Medicine, USF Health Heart Institute, MDD 0630, 560 Channelside Dr, Tampa, FL, 33602, USA
- University of South Florida, Morsani College of Medicine, Department of Molecular Pharmacology & Physiology, 12901 Bruce B Downs Blvd, Tampa, FL, 33612, USA
| | - Patrick M Kochanek
- Safar Center for Resuscitation Research, UPMC Children's Hospital of Pittsburgh, Rangos Research Center - 6th floor, Pittsburgh, PA, 15224, USA
| | - Travis C Jackson
- University of South Florida, Morsani College of Medicine, USF Health Heart Institute, MDD 0630, 560 Channelside Dr, Tampa, FL, 33602, USA.
- University of South Florida, Morsani College of Medicine, Department of Molecular Pharmacology & Physiology, 12901 Bruce B Downs Blvd, Tampa, FL, 33612, USA.
| |
Collapse
|
4
|
Gu Y, Chen X, Fu S, Liu W, Wang Q, Liu KJ, Shen J. Astragali Radix Isoflavones Synergistically Alleviate Cerebral Ischemia and Reperfusion Injury Via Activating Estrogen Receptor-PI3K-Akt Signaling Pathway. Front Pharmacol 2021; 12:533028. [PMID: 33692686 PMCID: PMC7937971 DOI: 10.3389/fphar.2021.533028] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 01/15/2021] [Indexed: 11/13/2022] Open
Abstract
Isoflavones are major neuroprotective components of a medicinal herb Astragali Radix, against cerebral ischemia-reperfusion injury but the mechanisms of neuroprotection remain unclear. Calycosin and formononetin are two major AR isoflavones while daidzein is the metabolite of formononetin after absorption. Herein, we aim to investigate the synergistic neuroprotective effects of those isoflavones of Astragali Radix against cerebral ischemia-reperfusion injury. Calycosin, formononetin and daidzein were organized with different combinations whose effects observed in both in vitro and in vivo experimental models. In the in vitro study, primary cultured neurons were subjected to oxygen-glucose deprivation plus reoxygenation (OGD/RO) or l-glutamate treatment. In the in vivo study, rats were subjected to middle cerebral artery occlusion to induce cerebral ischemia and reperfusion. All three isoflavones pre-treatment alone decreased brain infarct volume and improved neurological deficits in rats, and dose-dependently attenuated neural death induced by l-glutamate treatment and OGD/RO in cultured neurons. Interestingly, the combined formulas of those isoflavones revealed synergistically activated estrogen receptor (estrogen receptors)-PI3K-Akt signaling pathway. Using ER antagonist and phosphatidylinositol 3-kinase (PI3K) inhibitor blocked the neuroprotective effects of those isoflavones. In conclusion, isoflavones could synergistically alleviate cerebral ischemia-reperfusion injury via activating ER-PI3K-Akt pathway.
Collapse
Affiliation(s)
- Yong Gu
- Clinical Research Center, Hainan Provincial Hospital of Chinese Medicine, Haikou, China.,School of Chinese Medicine, University of Hong Kong, Hong Kong, China
| | - Xi Chen
- Department of Core Facility, The People's Hospital of Bao-an Shenzhen, Shenzhen, China.,School of Chinese Medicine, University of Hong Kong, Hong Kong, China
| | - Shuping Fu
- School of Chinese Medicine, University of Hong Kong, Hong Kong, China
| | - Wenlan Liu
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico, Albuquerque, NM, United States
| | - Qi Wang
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ke-Jian Liu
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico, Albuquerque, NM, United States
| | - Jiangang Shen
- School of Chinese Medicine, University of Hong Kong, Hong Kong, China.,The University of Hong Kong-Shenzhen, Institute of Research and Innovation (HKU-SIRI), Shenzhen, China
| |
Collapse
|
5
|
Tibolone Ameliorates the Lipotoxic Effect of Palmitic Acid in Normal Human Astrocytes. Neurotox Res 2020; 38:585-595. [DOI: 10.1007/s12640-020-00247-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 06/13/2020] [Accepted: 06/17/2020] [Indexed: 02/06/2023]
|
6
|
Intraventricular Medium B Treatment Benefits an Ischemic Stroke Rodent Model via Enhancement of Neurogenesis and Anti-apoptosis. Sci Rep 2020; 10:6596. [PMID: 32313130 PMCID: PMC7171187 DOI: 10.1038/s41598-020-63598-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 03/31/2020] [Indexed: 12/26/2022] Open
Abstract
Enhancement of endogenous neurogenesis after ischemic stroke may improve functional recovery. We previously demonstrated that medium B, which is a combination with epidermal growth factor (EGF) and fibronectin, can promote neural stem/progenitor cell (NSPC) proliferation and migration. Here, we showed that medium B promoted proliferation and migration of cultured NSPCs onto various 3-dimentional structures. When rat cortical neurons with oxygen glucose deprivation (OGD) were co-cultured with NSPCs, medium B treatment increased neuronal viability and reduced cell apoptosis. In a rat model with transient middle cerebral artery occlusion (MCAO), post-insult intraventricular medium B treatment enhanced proliferation, migration, and neuronal differentiation of NSPCs and diminished cell apoptosis in the infarct brain. In cultured post-OGD neuronal cells and the infarct brain from MCAO rats, medium B treatment increased protein levels of Bcl-xL, Bcl-2, phospho-Akt, phospho-GSK-3β, and β-catenin and decreased the cleaved caspase-3 level, which may be associated with the effects of anti-apoptosis. Notably, intraventricular medium B treatment increased neuronal density, improved motor function and reduced infarct size in MCAO rats. In summary, medium B treatment results in less neuronal death and better functional outcome in both cellular and rodent models of ischemic stroke, probably via promotion of neurogenesis and reduction of apoptosis.
Collapse
|
7
|
Ishihara Y, Sakurai H, Oguro A, Tsuji M, Vogel CFA, Yamazaki T. Retinoid X receptor-mediated neuroprotection via CYP19 upregulation and subsequent increases in estradiol synthesis. J Steroid Biochem Mol Biol 2019; 193:105421. [PMID: 31265900 DOI: 10.1016/j.jsbmb.2019.105421] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 06/14/2019] [Accepted: 06/28/2019] [Indexed: 10/26/2022]
Abstract
Increasing evidence has shown that one of the major neurosteroids, estradiol, has potent neuroprotective actions. We have reported that estradiol synthesis was enhanced when retinoic acid was added into rat hippocampal slice culture. In this study, we investigated the effects of a potent retinoid X receptor (RXR) agonist, bexarotene, on estrogen synthesis and neuroprotective action in hippocampal slices. Treatment with bexarotene increased estradiol levels as well as estrogen-synthesizing enzymes and CYP19 expression in hippocampal slice cultures. Bexarotene significantly suppressed neuronal cell death induced by oxygen-glucose deprivation (OGD)/reoxygenation. RXR agonists other than bexarotene, such as CD3254, also suppressed neuronal cell death accompanied by OGD/reoxygenation. The RXR antagonists HX531 and UVI3003 and the CYP19 inhibitor letrozole abolished the neuroprotection elicited by bexarotene, indicating that estradiol produced by RXR stimulation protects neurons from ischemic insult. The human brain-specific CYP19 promoter had 6 RXR half sites, and 2 of 6 half sites were responsible for CYP19 expression induced by bexarotene. Bexarotene increased the expression of catalase and glutathione peroxidase 1 and inhibited lipid peroxidation elicited by OGD/reoxygenation, suggesting that the antioxidative property of estrogen contributes to RXR-mediated neuroprotection. Bexarotene also suppressed neuronal injury induced by lipopolysaccharide in the hippocampal slices. Taken together, RXR stimulation can protect neurons via enhanced synthesis of estradiol with antioxidative mechanisms. The RXR-estrogen axis might be a novel mechanism-based strategy to prevent or ameliorate ischemic and/or inflammatory neuronal disorders.
Collapse
Affiliation(s)
- Yasuhiro Ishihara
- Program of Biomedical Science, Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima 739-8521, Japan; Center for Health and the Environment, University of California, Davis, CA, 95616, USA.
| | - Hikaru Sakurai
- Program of Biomedical Science, Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima 739-8521, Japan
| | - Ami Oguro
- Program of Biomedical Science, Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima 739-8521, Japan
| | - Mayumi Tsuji
- Department of Environmental Health, University of Occupational and Environmental Health, Fukuoka 807-8555, Japan
| | - Christoph F A Vogel
- Center for Health and the Environment, University of California, Davis, CA, 95616, USA; Department of Environmental Toxicology, University of California, Davis, CA, 95616, USA
| | - Takeshi Yamazaki
- Program of Life and Environmental Sciences, Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima 739-8521, Japan
| |
Collapse
|
8
|
Baez-Jurado E, Rincón-Benavides MA, Hidalgo-Lanussa O, Guio-Vega G, Ashraf GM, Sahebkar A, Echeverria V, Garcia-Segura LM, Barreto GE. Molecular mechanisms involved in the protective actions of Selective Estrogen Receptor Modulators in brain cells. Front Neuroendocrinol 2019; 52:44-64. [PMID: 30223003 DOI: 10.1016/j.yfrne.2018.09.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 09/09/2018] [Accepted: 09/12/2018] [Indexed: 02/06/2023]
Abstract
Synthetic selective modulators of the estrogen receptors (SERMs) have shown to protect neurons and glial cells against toxic insults. Among the most relevant beneficial effects attributed to these compounds are the regulation of inflammation, attenuation of astrogliosis and microglial activation, prevention of excitotoxicity and as a consequence the reduction of neuronal cell death. Under pathological conditions, the mechanism of action of the SERMs involves the activation of estrogen receptors (ERs) and G protein-coupled receptor for estrogens (GRP30). These receptors trigger neuroprotective responses such as increasing the expression of antioxidants and the activation of kinase-mediated survival signaling pathways. Despite the advances in the knowledge of the pathways activated by the SERMs, their mechanism of action is still not entirely clear, and there are several controversies. In this review, we focused on the molecular pathways activated by SERMs in brain cells, mainly astrocytes, as a response to treatment with raloxifene and tamoxifen.
Collapse
Affiliation(s)
- E Baez-Jurado
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá D.C., Colombia
| | - M A Rincón-Benavides
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá D.C., Colombia
| | - O Hidalgo-Lanussa
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá D.C., Colombia
| | - G Guio-Vega
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá D.C., Colombia
| | - G M Ashraf
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - A Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - V Echeverria
- Universidad San Sebastián, Fac. Cs de la Salud, Lientur 1457, Concepción 4080871, Chile; Research & Development Service, Bay Pines VA Healthcare System, Bay Pines, FL 33744, USA
| | - L M Garcia-Segura
- Instituto Cajal, CSIC, Madrid, Spain; Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - G E Barreto
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá D.C., Colombia; Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Santiago, Chile.
| |
Collapse
|
9
|
Pottoo FH, Javed MN, Barkat MA, Alam MS, Nowshehri JA, Alshayban DM, Ansari MA. Estrogen and Serotonin: Complexity of Interactions and Implications for Epileptic Seizures and Epileptogenesis. Curr Neuropharmacol 2019; 17:214-231. [PMID: 29956631 PMCID: PMC6425080 DOI: 10.2174/1570159x16666180628164432] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2018] [Revised: 02/01/2018] [Accepted: 06/25/2018] [Indexed: 12/15/2022] Open
Abstract
A burgeoning literature documents the confluence of ovarian steroids and central serotonergic systems in the injunction of epileptic seizures and epileptogenesis. Estrogen administration in animals reduces neuronal death from seizures by up-regulation of the prosurvival molecule i.e. Bcl-2, anti-oxidant potential and protection of NPY interneurons. Serotonin modulates epileptiform activity in either direction i.e administration of 5-HT agonists or reuptake inhibitors leads to the activation of 5-HT3 and 5-HT1A receptors tending to impede focal and generalized seizures, while depletion of brain 5-HT along with the destruction of serotonergic terminals leads to expanded neuronal excitability hence abatement of seizure threshold in experimental animal models. Serotonergic neurotransmission is influenced by the organizational activity of steroid hormones in the growing brain and the actuation effects of steroids which come in adulthood. It is further established that ovarian steroids bring induction of dendritic spine proliferation on serotonin neurons thus thawing a profound effect on serotonergic transmission. This review features 5-HT1A and 5-HT3 receptors as potential targets for ameliorating seizure-induced neurodegeneration and recurrent hypersynchronous neuronal activity. Indeed 5-HT3 receptors mediate cross-talk between estrogenic and serotonergic pathways, and could be well exploited for combinatorial drug therapy against epileptogenesis.
Collapse
Affiliation(s)
- Faheem Hyder Pottoo
- Address correspondence to these authors at the Department of Epidemic Disease Research, Institute of Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University (Formerly University of Dammam), Dammam 31441, Saudi Arabia; E-mail: and Department of Pharmacology, College of Clinical Pharmacy, 31441 Imam Abdulrahman Bin Faisal University, (Formerly University of Dammam), Dammam, Saudi Arabia; E-mail:
| | | | | | | | | | | | - Mohammad Azam Ansari
- Address correspondence to these authors at the Department of Epidemic Disease Research, Institute of Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University (Formerly University of Dammam), Dammam 31441, Saudi Arabia; E-mail: and Department of Pharmacology, College of Clinical Pharmacy, 31441 Imam Abdulrahman Bin Faisal University, (Formerly University of Dammam), Dammam, Saudi Arabia; E-mail:
| |
Collapse
|
10
|
Baidukova O, Wang Q, Chaiwaree S, Freyer D, Prapan A, Georgieva R, Zhao L, Bäumler H. Antioxidative protection of haemoglobin microparticles (HbMPs) by PolyDopamine. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2018; 46:S693-S701. [PMID: 30450978 DOI: 10.1080/21691401.2018.1505748] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Clinically applicable haemoglobin-based oxygen carriers (HBOCs) should neither induce immunological nor toxic reactions. Additionally, Hb should be protected against oxidation. In the absence of protective enzymes (superoxide dismutase (SOD) and catalase (CAT)) Hb is oxidized to MetHb and thus losing its function of oxygen delivery. Alternatively, polydopamine (PD), a scavenger of free radicals, could be used for Hb protection against oxidation Therefore, we synthetized HbMPs modified with PD. The content of functional haemoglobin in these PD-HbMPs was twice higher than that in the control HbMPs due to the protective antioxidant effect of PD. In addition, the PD-HbMPs exhibited a high scavenging activity of free radicals including H2O2 and excellent biocompatibility. In contrast to monomeric dopamine, which has been shown to produce toxic effects on neurons due to formation of H2O2, hydroxyl radicals and superoxide during the process of auto-oxidation, PD-HbMPs are not neurotoxic. Consequently, the results presented here suggest a great potential of PD-HbMPs as HBOCs.
Collapse
Affiliation(s)
- Olga Baidukova
- a Charité-Universitätsmedizin Berlin, Institute of Transfusion Medicine , Berlin , Germany
| | - Quan Wang
- a Charité-Universitätsmedizin Berlin, Institute of Transfusion Medicine , Berlin , Germany.,b Institute of Health Service and Transfusion Medicine , Bejing , PR China
| | - Saranya Chaiwaree
- a Charité-Universitätsmedizin Berlin, Institute of Transfusion Medicine , Berlin , Germany.,c Faculty of Pharmacy , Payap University , Chiang Mai , Thailand
| | - Dorette Freyer
- d Department of Experimental Neurology, Clinic of Neurology , Charité-Universitätsmedizin Berlin , Berlin , Germany
| | - Ausanai Prapan
- a Charité-Universitätsmedizin Berlin, Institute of Transfusion Medicine , Berlin , Germany.,e Department of Radiological Technology , Naresuan University , Phitsanulok , Thailand
| | - Radostina Georgieva
- a Charité-Universitätsmedizin Berlin, Institute of Transfusion Medicine , Berlin , Germany.,f Medical Faculty, Department of Medical Physics, Biophysics, and Radiology , Trakia Unversity , Stara Zagora , Bulgaria
| | - Lian Zhao
- b Institute of Health Service and Transfusion Medicine , Bejing , PR China
| | - Hans Bäumler
- a Charité-Universitätsmedizin Berlin, Institute of Transfusion Medicine , Berlin , Germany
| |
Collapse
|
11
|
Stelmashook EV, Genrikhs EE, Kapkaeva MR, Zelenova EA, Isaev NK. N-Acetyl-L-cysteine in the Presence of Cu 2+ Induces Oxidative Stress and Death of Granule Neurons in Dissociated Cultures of Rat Cerebellum. BIOCHEMISTRY (MOSCOW) 2017; 82:1176-1182. [PMID: 29037138 DOI: 10.1134/s0006297917100108] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Addition into the culture medium of the antioxidant N-acetylcysteine (NAC, 1 mM) in the presence of Cu2+ (0.0005-0.001 mM) induced intensive death of cultured rat cerebellar granule neurons, which was significantly decreased by the zinc ion chelator TPEN (N,N,N',N'-tetrakis(2-pyridylmethyl)ethylenediamine). However, the combined action of NAC and Zn2+ did not induce destruction of the neurons. Measurement of the relative intracellular concentration of Zn2+ with the fluorescent probe FluoZin-3 AM or of free radical production using a CellROX Green showed that incubation of the culture for 4 h with Cu2+ and NAC induced an intensive increase in the fluorescence of CellROX Green but not of FluoZin-3. Probably, the protective effect of TPEN in this case could be mediated by its ability to chelate Cu2+. Incubation of cultures in a balanced salt solution in the presence of 0.01 mM Cu2+ caused neuronal death already after 1 h if the NAC concentration in the solution was within 0.005-0.05 mM. NAC at higher concentrations (0.1-1 mM) together with 0.01 mM Cu2+ did not cause the death of neurons. These data imply that the antioxidant NAC can be potentially harmful to neurons even in the presence of nanomolar concentrations of variable valence metals.
Collapse
|
12
|
Engler-Chiurazzi EB, Brown CM, Povroznik JM, Simpkins JW. Estrogens as neuroprotectants: Estrogenic actions in the context of cognitive aging and brain injury. Prog Neurobiol 2017; 157:188-211. [PMID: 26891883 PMCID: PMC4985492 DOI: 10.1016/j.pneurobio.2015.12.008] [Citation(s) in RCA: 146] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 11/06/2015] [Accepted: 12/10/2015] [Indexed: 12/30/2022]
Abstract
There is ample empirical evidence to support the notion that the biological impacts of estrogen extend beyond the gonads to other bodily systems, including the brain and behavior. Converging preclinical findings have indicated a neuroprotective role for estrogen in a variety of experimental models of cognitive function and brain insult. However, the surprising null or even detrimental findings of several large clinical trials evaluating the ability of estrogen-containing hormone treatments to protect against age-related brain changes and insults, including cognitive aging and brain injury, led to hesitation by both clinicians and patients in the use of exogenous estrogenic treatments for nervous system outcomes. That estrogen-containing therapies are used by tens of millions of women for a variety of health-related applications across the lifespan has made identifying conditions under which benefits with estrogen treatment will be realized an important public health issue. Here we provide a summary of the biological actions of estrogen and estrogen-containing formulations in the context of aging, cognition, stroke, and traumatic brain injury. We have devoted special attention to highlighting the notion that estrogen appears to be a conditional neuroprotectant whose efficacy is modulated by several interacting factors. By developing criteria standards for desired beneficial peripheral and neuroprotective outcomes among unique patient populations, we can optimize estrogen treatments for attenuating the consequences of, and perhaps even preventing, cognitive aging and brain injury.
Collapse
Affiliation(s)
- E B Engler-Chiurazzi
- Center for Basic and Translational Stroke Research, West Virginia University, Morgantown, WV 26506, United States; Department of Physiology and Pharmacology, West Virginia University, Morgantown, WV 26506, United States.
| | - C M Brown
- Center for Basic and Translational Stroke Research, West Virginia University, Morgantown, WV 26506, United States; Department of Neurobiology and Anatomy, West Virginia University, Morgantown, WV 26506, United States.
| | - J M Povroznik
- Center for Basic and Translational Stroke Research, West Virginia University, Morgantown, WV 26506, United States; Department of Pediatrics, West Virginia University, Morgantown, WV 26506, United States.
| | - J W Simpkins
- Center for Basic and Translational Stroke Research, West Virginia University, Morgantown, WV 26506, United States; Department of Physiology and Pharmacology, West Virginia University, Morgantown, WV 26506, United States.
| |
Collapse
|
13
|
Posod A, Wechselberger K, Stanika RI, Obermair GJ, Wegleiter K, Huber E, Urbanek M, Kiechl-Kohlendorfer U, Griesmaier E. Administration of secretoneurin is protective in hypoxic-ischemic neonatal brain injury predominantly in the hypoxic-only hemisphere. Neuroscience 2017; 352:88-96. [PMID: 28391015 DOI: 10.1016/j.neuroscience.2017.03.055] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2017] [Revised: 03/27/2017] [Accepted: 03/28/2017] [Indexed: 10/19/2022]
Abstract
Neonatal brain injury is a problem of global importance. To date, no causal therapies are available. A substance with considerable therapeutic potential is the endogenous neuropeptide secretoneurin (SN), which has proven to be beneficial in adult stroke. The aim of this study was to assess its effect in neonatal hypoxic-ischemic brain injury models. In vitro, primary hippocampal neurons were pre-treated with vehicle, 1µg/ml, 10µg/ml, or 50µg/ml SN and subjected to oxygen-glucose deprivation (OGD) for six hours. Cell death was assessed after a 24-h recovery period. In vivo, seven day-old CD-1 mice underwent unilateral common carotid artery ligation and were exposed to 8% oxygen/nitrogen for 20 min. SN plasma concentrations were serially determined by ELISA after insult. One hour after hypoxia, a subgroup of animals was treated with vehicle or SN. SN plasma concentrations significantly decreased 48h after insult. The number of caspase-3-positive cells was significantly lower in the hypoxic-ischemic hemisphere in the thalamus of SN-treated animals. In the hypoxic-only hemisphere administration of SN significantly reduced the number of caspase-3-positive cells (in cortex, white matter, hippocampus, thalamus and striatum) and inhibited microglial cell activation in the thalamus. SN has neuroprotective potential in neonatal brain injury. Its main action seems to be inhibition of apoptosis in the aftermath of the insult, predominantly in the hypoxic-only hemisphere. This might be explained by the less pronounced injury in this hemisphere, where blood flow and thus nutrient supply are maintained.
Collapse
Affiliation(s)
- Anna Posod
- Pediatrics II (Neonatology), Department of Pediatrics, Medical University of Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria
| | - Karina Wechselberger
- Pediatrics II (Neonatology), Department of Pediatrics, Medical University of Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria
| | - Ruslan Iljitsch Stanika
- Division of Physiology, Department of Physiology and Medical Physics, Medical University of Innsbruck, Schöpfstrasse 41, 6020 Innsbruck, Austria
| | - Gerald J Obermair
- Division of Physiology, Department of Physiology and Medical Physics, Medical University of Innsbruck, Schöpfstrasse 41, 6020 Innsbruck, Austria
| | - Karina Wegleiter
- Pediatrics II (Neonatology), Department of Pediatrics, Medical University of Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria
| | - Eva Huber
- Pediatrics II (Neonatology), Department of Pediatrics, Medical University of Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria
| | - Martina Urbanek
- Pediatrics II (Neonatology), Department of Pediatrics, Medical University of Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria
| | - Ursula Kiechl-Kohlendorfer
- Pediatrics II (Neonatology), Department of Pediatrics, Medical University of Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria
| | - Elke Griesmaier
- Pediatrics II (Neonatology), Department of Pediatrics, Medical University of Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria.
| |
Collapse
|
14
|
Wnuk A, Rzemieniec J, Lasoń W, Krzeptowski W, Kajta M. Apoptosis Induced by the UV Filter Benzophenone-3 in Mouse Neuronal Cells Is Mediated via Attenuation of Erα/Pparγ and Stimulation of Erβ/Gpr30 Signaling. Mol Neurobiol 2017; 55:2362-2383. [PMID: 28357806 PMCID: PMC5840254 DOI: 10.1007/s12035-017-0480-z] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Accepted: 03/03/2017] [Indexed: 12/22/2022]
Abstract
Although benzophenone-3 (BP-3) has frequently been reported to play a role in endocrine disruption, there is insufficient data regarding the impact of BP-3 on the nervous system, including its possible adverse effects on the developing brain. Our study demonstrated that BP-3 caused neurotoxicity and activated apoptosis via an intrinsic pathway involving the loss of mitochondrial membrane potential and the activation of caspases-9 and -3 and kinases p38/MAPK and Gsk3β. These biochemical alterations were accompanied by ROS production, increased apoptotic body formation and impaired cell survival, and by an upregulation of the genes involved in apoptosis. The BP-3-induced effects were tissue-specific and age-dependent with the most pronounced effects observed in neocortical cells at 7 days in vitro. BP-3 changed the messenger RNA (mRNA) expression levels of Erα, Erβ, Gpr30, and Pparγ in a time-dependent manner. At 3 h of exposure, BP-3 downregulated estrogen receptor mRNAs but upregulated Pparγ mRNA. After prolonged exposures, BP-3 downregulated the receptor mRNAs except for Erβ mRNA that was upregulated. The BP-3-induced patterns of mRNA expression measured at 6 and 24 h of exposure reflected alterations in the protein levels of the receptors and paralleled their immunofluorescent labeling. Erα and Pparγ agonists diminished, but Erβ and Gpr30 agonists stimulated the BP-3-induced apoptotic and neurotoxic effects. Receptor antagonists caused the opposite effects, except for ICI 182,780. This is in line with a substantial reduction in the effects of BP-3 in cells with siRNA-silenced Erβ/Gpr30 and the maintenance of BP-3 effects in Erα- and Pparγ siRNA-transfected cells. We showed for the first time that BP-3-affected mRNA and protein expression levels of Erα, Erβ, Gpr30, and Pparγ, paralleled BP-3-induced apoptosis and neurotoxicity. Therefore, we suggest that BP-3-evoked apoptosis of neuronal cells is mediated via attenuation of Erα/Pparγ and stimulation of Erβ/Gpr30 signaling.
Collapse
Affiliation(s)
- A Wnuk
- Department of Experimental Neuroendocrinology, Institute of Pharmacology, Polish Academy of Sciences, Smetna Street 12, 31-343, Krakow, Poland
| | - J Rzemieniec
- Department of Experimental Neuroendocrinology, Institute of Pharmacology, Polish Academy of Sciences, Smetna Street 12, 31-343, Krakow, Poland
| | - W Lasoń
- Department of Experimental Neuroendocrinology, Institute of Pharmacology, Polish Academy of Sciences, Smetna Street 12, 31-343, Krakow, Poland
| | - W Krzeptowski
- Department of Cell Biology and Imaging, Institute of Zoology, Jagiellonian University, Gronostajowa Street 9, 30-387, Krakow, Poland
| | - M Kajta
- Department of Experimental Neuroendocrinology, Institute of Pharmacology, Polish Academy of Sciences, Smetna Street 12, 31-343, Krakow, Poland.
| |
Collapse
|
15
|
Domin H, Przykaza Ł, Jantas D, Kozniewska E, Boguszewski PM, Śmiałowska M. Neuroprotective potential of the group III mGlu receptor agonist ACPT-I in animal models of ischemic stroke: In vitro and in vivo studies. Neuropharmacology 2016; 102:276-94. [DOI: 10.1016/j.neuropharm.2015.11.025] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Revised: 11/07/2015] [Accepted: 11/24/2015] [Indexed: 01/21/2023]
|
16
|
Domin H, Jantas D, Śmiałowska M. Neuroprotective effects of the allosteric agonist of metabotropic glutamate receptor 7 AMN082 on oxygen-glucose deprivation- and kainate-induced neuronal cell death. Neurochem Int 2015; 88:110-23. [PMID: 25576184 DOI: 10.1016/j.neuint.2014.12.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Revised: 12/07/2014] [Accepted: 12/17/2014] [Indexed: 12/24/2022]
Abstract
Although numerous studies demonstrated a neuroprotective potency of unspecific group III mGluR agonists in in vitro and in vivo models of excitotoxicity, little is known about the protective role of group III mGlu receptor activation against neuronal cell injury evoked by ischemic conditions. The aim of the present study was to assess neuroprotective potential of the allosteric agonist of mGlu7 receptor, N,N'-Bis(diphenylmethyl)-1,2-ethanediamine dihydrochloride (AMN082) against oxygen-glucose deprivation (OGD)- and kainate (KA)-evoked neuronal cell damage in primary neuronal cultures, with special focus on its efficacy after delayed application. We demonstrated that in cortical neuronal cultures exposed to a 180 min OGD, AMN082 (0.01-1 µM) in a concentration- and time-dependent way attenuated the OGD-induced changes in the LDH release and MTT reduction assays. AMN082 (0.5 and 1 µM) produced also neuroprotective effects against KA-evoked neurotoxicity both in cortical and hippocampal cultures. Of particular importance was the finding that AMN082 attenuated excitotoxic neuronal injury after delayed application (30 min after OGD, or 30 min-1 h after KA). In both models of neurotoxicity, namely OGD- and KA-induced injury, the neuroprotective effects of AMN082 (1 µM) were reversed by the selective mGlu7 antagonist, 6-(4-Methoxyphenyl)-5-methyl-3-(4-pyridinyl)-isoxazolo[4,5-c]pyridin-4(5H)-one hydrochloride (MMPIP, 1 µM), suggesting the mGlu7-dependent mechanism of neuroprotective effects of AMN082. Next, we showed that AMN082 (0.5 and 1 µM) attenuated the OGD-induced increase in the number of necrotic nuclei as well inhibited the OGD-evoked calpain activation, suggesting the participation of these processes in the mechanism of AMN082-mediated protection. Additionally, we showed that protection evoked by AMN082 (1 µM) in KA model was connected with the inhibition of toxin-induced caspase-3 activity, and this effect was abolished by the mGlu7 receptor antagonist. The obtained results indicated that the activation of mGlu7 receptors may be a promising target for neuroprotection against ischemic and excitotoxic insults.
Collapse
Affiliation(s)
- Helena Domin
- Department of Neurobiology, Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343 Kraków, Poland.
| | - Danuta Jantas
- Department of Experimental Neuroendocrinology, Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343 Kraków, Poland
| | - Maria Śmiałowska
- Department of Neurobiology, Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343 Kraków, Poland
| |
Collapse
|
17
|
Liu H, Yang L, Zhao Y, Zeng G, Wu Y, Chen Y, Zhang J, Zeng Q. Estrogen is a novel regulator of Tnfaip1 in mouse hippocampus. Int J Mol Med 2014; 34:219-27. [PMID: 24737445 DOI: 10.3892/ijmm.2014.1742] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2013] [Accepted: 02/26/2014] [Indexed: 11/06/2022] Open
Abstract
Tumor necrosis factor‑induced protein 1 (Tnfaip1), also known as B12, has been previously identified as a tumor necrosis factor-α (TNF-α)-inducible protein and is involved in the cytokinesis signaling pathway, DNA synthesis, innate immunity, cell apoptosis, Alzheimer's disease (AD) and type 2 diabetic nephropathy. However, little is known regarding the expression of Tnfaip1 in various tissues or its accurate role in these physiological functions. The focus of this study was on Tnfaip1 expression in different tissues, with a high expression in mouse hippocampus being identified. The age- and gender‑related expression of Tnfaip1 in hippocampus was also investigated. The distribution of Tnfaip1 was mapped using fluorescent immunostaining. Although immunoactivity was found in the CA1, CA3 and DG subregions of the hippocampus in E17.5 and P6 mice, strong staining was only detected in the CA3 subregion in adult mice. These data suggested that Tnfaip1 expression in hippocampus may be regulated by estrogen. Further study showed that the expression of Tnfaip1 in the hippocampus was significantly increased in ovariecto-mized mice compared to Sham mice. In cultured primary hippocampal cells, Tnfaip1 showed different expression levels in different treatments of estrogen or estrogen receptor antagonists. Additional experiments demonstrated the existence of a binding site of ERβ in the Tnfaip1 promoter region, and that ERβ was able to upregulate Tnfaip1 expression. Our study identified a new regulatory factor and a primary regulatory mechanism of Tnfaip1 expression in hippocampus. Since both hippocampus and estrogen are crucial in AD, the results also showed a potential association between Tnfaip1 and hippocampal-related diseases, such as AD, which may be affected by the estrogen level.
Collapse
Affiliation(s)
- Hui Liu
- Department of Life Science, Hunan Agricultural University, Changsha, Hunan 410128, P.R. China
| | - Liping Yang
- Department of Enviromental Science, Changsha Environmental Protection College, Changsha, Hunan 410004, P.R. China
| | - Yingchun Zhao
- Department of Biostatistics and Bioinformatics, Tulane University, New Orleans, LA 70112, USA
| | - Guihua Zeng
- Department of Enviromental Science, Changsha Environmental Protection College, Changsha, Hunan 410004, P.R. China
| | - Yaosong Wu
- Laboratory of Molecular Biology, Henan University of Traditional Chinese Medicine, Zhengzhou, Henan 410208, P.R. China
| | - Yulong Chen
- Laboratory of Molecular Biology, Henan University of Traditional Chinese Medicine, Zhengzhou, Henan 410208, P.R. China
| | - Jian Zhang
- Key Laboratory of Protein Chemistry and Developmental Biology, Ministry of Education of China, Department of Biochemistry and Molecular Biology, College of Life Science, Hunan Normal University, Changsha, Hunan 410081, P.R. China
| | - Qingru Zeng
- Department of Life Science, Hunan Agricultural University, Changsha, Hunan 410128, P.R. China
| |
Collapse
|
18
|
Marin R, Casañas V, Pérez JA, Fabelo N, Fernandez CE, Diaz M. Oestrogens as modulators of neuronal signalosomes and brain lipid homeostasis related to protection against neurodegeneration. J Neuroendocrinol 2013; 25:1104-15. [PMID: 23795744 DOI: 10.1111/jne.12068] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Revised: 05/22/2013] [Accepted: 06/18/2013] [Indexed: 12/19/2022]
Abstract
Oestrogens trigger several pathways at the plasma membrane that exert beneficial actions against neurodegenerative diseases, such as Alzheimer's disease and Parkinson's disease. Part of these actions takes place in lipid rafts, which are membrane domains with a singular protein and lipid composition. These microdomains also represent a preferential site for signalling protein complexes, or signalosomes. A plausible hypothesis is that the dynamic interaction of signalosomes with different extracellular ligands may be at the basis of neuronal maintenance against different neuropathologies. Oestrogen receptors are localised in neuronal lipid rafts, taking part of macromolecular complexes together with a voltage-dependent anion channel (VDAC), and other molecules. Oestradiol binding to its receptor at this level enhances neuroprotection against amyloid-β degeneration through the activation of different signal transduction pathways, including VDAC gating modulation. Moreover, part of the stability and functionality of signalling platforms lays on the distribution of lipid hallmarks in these microstructures, which modulate membrane physicochemical properties, thus favouring molecular interactions. Interestingly, recent findings indicate a potential role of oestrogens in the preservation of neuronal membrane physiology related to lipid homeostasis. Thus, oestrogens and docosahexaenoic acid may act synergistically to stabilise brain lipid structure by regulating neuronal lipid biosynthetic pathways, suggesting that part of the neuroprotective effects elicited by oestrogens occur through mechanisms aimed at preserving lipid homeostasis. Overall, oestrogen mechanisms of neuroprotection may occur not only by its interaction with neuronal protein targets through nongenomic and genomic mechanisms, but also through its participation in membrane architecture stabilisation via 'lipostatic' mechanisms.
Collapse
Affiliation(s)
- R Marin
- Department of Physiology, Laboratory of Cellular Neurobiology, University of La Laguna, La Laguna, Tenerife, Spain
| | | | | | | | | | | |
Collapse
|
19
|
Martins JB, Bastos MDL, Carvalho F, Capela JP. Differential Effects of Methyl-4-Phenylpyridinium Ion, Rotenone, and Paraquat on Differentiated SH-SY5Y Cells. J Toxicol 2013; 2013:347312. [PMID: 23710172 PMCID: PMC3654346 DOI: 10.1155/2013/347312] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2012] [Revised: 01/28/2013] [Accepted: 01/28/2013] [Indexed: 12/21/2022] Open
Abstract
Paraquat (PQ), a cationic nonselective bipyridyl herbicide, has been used as neurotoxicant to modulate Parkinson's disease in laboratory settings. Other compounds like rotenone (ROT), a pesticide, and 1-methyl-4-phenylpyridinium ion (MPP(+)) have been widely used as neurotoxicants. We compared the toxicity of these three neurotoxicants using differentiated dopaminergic SH-SY5Y human cells, aiming to elucidate their differential effects. PQ-induced neurotoxicity was shown to be concentration and time dependent, being mitochondrial dysfunction followed by neuronal death. On the other hand, cells exposure to MPP(+) induced mitochondrial dysfunction, but not cellular lyses. Meanwhile, ROT promoted both mitochondrial dysfunction and neuronal death, revealing a biphasic pattern. To further elucidate PQ neurotoxic mechanism, several protective agents were used. SH-SY5Y cells pretreatment with tiron (TIR) and 2-hydroxybenzoic acid sodium salt (NaSAL), both antioxidants, and N ω -nitro-L-arginine methyl ester hydrochloride (L-NAME), a nitric oxide synthase inhibitor, partially protected against PQ-induced cell injury. Additionally, 1-(2-[bis(4-fluorophenyl)methoxy]ethyl)-4-(3-phenyl-propyl)piperazine (GBR 12909), a dopamine transporter inhibitor, and cycloheximide (CHX), a protein synthesis inhibitor, also partially protected against PQ-induced cell injury. In conclusion, we demonstrated that PQ, MPP(+), and ROT exerted differential toxic effects on dopaminergic cells. PQ neurotoxicity occurred through exacerbated oxidative stress, with involvement of uptake through the dopamine transporter and protein synthesis.
Collapse
Affiliation(s)
- João Barbosa Martins
- REQUIMTE (Rede de Química e Tecnologia), Laboratório de Toxicologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Maria de Lourdes Bastos
- REQUIMTE (Rede de Química e Tecnologia), Laboratório de Toxicologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Félix Carvalho
- REQUIMTE (Rede de Química e Tecnologia), Laboratório de Toxicologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - João Paulo Capela
- REQUIMTE (Rede de Química e Tecnologia), Laboratório de Toxicologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
- Faculty of Health Sciences, University Fernando Pessoa, Rua Carlos da Maia 296, 4200-150 Porto, Portugal
| |
Collapse
|
20
|
Campos FL, Cristovão AC, Rocha SM, Fonseca CP, Baltazar G. GDNF contributes to oestrogen-mediated protection of midbrain dopaminergic neurones. J Neuroendocrinol 2012; 24:1386-97. [PMID: 22672424 DOI: 10.1111/j.1365-2826.2012.02348.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Parkinson's disease (PD) is characterised by the preferential loss of dopaminergic neurones from the substantia nigra (SN) that leads to the hallmark motor disturbances. Animal and human studies suggest a beneficial effect of oestrogen to the nigrostriatal system, and the regulation of neurotrophic factor expression by oestrogens has been suggested as a possible mechanism contributing to that neuroprotective effect. The present study was designed to investigate whether the neuroprotection exerted by 17β-oestradiol on nigrostriatal dopaminergic neurones is mediated through the regulation of glial cell line-derived neurotrophic factor (GDNF) expression. Using an in vivo rat model of PD, we were able to confirm the relevance of 17β-oestradiol in defending dopaminergic neurones against 6-hydroxydopamine (6-OHDA) toxicity. 17β-oestradiol, released by micro-osmotic pumps, implanted 10 days before intrastriatal 6-OHDA injection, prevented the loss of dopaminergic neurones induced by 6-OHDA. 17β-oestradiol treatment also promoted an increase in GDNF protein levels both in the SN and striatum. To explore the relevance of GDNF increases to 17β-oestradiol neuroprotection, we analysed, in SN neurone-glia cultures, the effect of GDNF antibody neutralisation and RNA interference-mediated GDNF knockdown. The results showed that both GDNF neutralisation and GDNF silencing abolished the dopaminergic protection provided by 17β-oestradiol against 6-OHDA toxicity. Taken together, these results strongly identify GDNF as an important player in 17β-oestradiol-mediated dopaminergic neuroprotection.
Collapse
Affiliation(s)
- F L Campos
- CICS-UBI - Health Sciences Research Centre, Department of Medical Sciences, University of Beira Interior, Covilhã, Portugal
| | | | | | | | | |
Collapse
|
21
|
Bayer J, Rune G, Kutsche K, Schwarze U, Kalisch R, Büchel C, Sommer T. Estrogen and the male hippocampus: Genetic variation in the aromatase gene predicting serum estrogen is associated with hippocampal gray matter volume in men. Hippocampus 2012; 23:117-21. [DOI: 10.1002/hipo.22059] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/10/2012] [Indexed: 01/11/2023]
|
22
|
Spampinato SF, Molinaro G, Merlo S, Iacovelli L, Caraci F, Battaglia G, Nicoletti F, Bruno V, Sortino MA. Estrogen receptors and type 1 metabotropic glutamate receptors are interdependent in protecting cortical neurons against β-amyloid toxicity. Mol Pharmacol 2012; 81:12-20. [PMID: 21984253 DOI: 10.1124/mol.111.074021] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/14/2025] Open
Abstract
We examined the interaction between estrogen receptors (ERs) and type 1 metabotropic glutamate receptors (mGlu1 receptors) in mechanisms of neurodegeneration/neuroprotection using mixed cultures of cortical cells challenged with β-amyloid peptide. Both receptors were present in neurons, whereas only ERα but not mGlu1 receptors were found in astrocytes. Addition of 17β-estradiol (17βE2) protected cultured neurons against amyloid toxicity, and its action was mimicked by the selective ERα agonist, 1,3,5-tris(4-hydroxyphenyl)-4-propyl-1H-pyrazole (PPT) as well as by a cell-impermeable bovine serum albumin conjugate of 17βE2. The selective ERβ agonist, diarylpropionitrile (DPN), was only slightly neuroprotective. The mGlu1/5 receptor agonist, 3,5-dihydroxyphenylglycine (DHPG), was also neuroprotective against amyloid toxicity, and its action was abolished by the mGlu1 receptor antagonist, (3,4-dihydro-2H-pyrano[2,3-b]quinolin-7-yl)-(cis-4-methoxycyclohexyl)-methanone (JNJ 16259685). Neuroprotection by 17βΕ2 or PPT (but not DPN) and DHPG was less than additive, suggesting that ERα and mGlu1 receptors activate the same pathway of cell survival. More important, neuroprotection by 17βΕ2 was abolished not only by the ER antagonist fulvestrant (ICI 182,780) but also by JNJ 16259685, and neuroprotection by DHPG was abolished by ICI 182,780. ERα and mGlu1 receptors were also interdependent in activating the phosphatidylinositol-3-kinase pathway, and pharmacological blockade of this pathway abolished neuroprotection by 17βE2, DHPG, or their combination. These data provide the first evidence that ERα and mGlu1 receptors critically interact in promoting neuroprotection, information that should be taken into account when the impact of estrogen on neurodegeneration associated with central nervous system disorders is examined.
Collapse
|
23
|
Datwyler AL, Lättig-Tünnemann G, Yang W, Paschen W, Lee SLL, Dirnagl U, Endres M, Harms C. SUMO2/3 conjugation is an endogenous neuroprotective mechanism. J Cereb Blood Flow Metab 2011; 31:2152-9. [PMID: 21863037 PMCID: PMC3210338 DOI: 10.1038/jcbfm.2011.112] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Small ubiquitin-like modifier (SUMO)2/3 but not SUMO1 conjugation is activated after transient cerebral ischemia. To investigate its function, we blocked neuronal SUMO2/3 translation through lentiviral microRNA delivery in primary cortical neurons. Viability was unaffected by SUMO2/3 silencing unless neurons were stressed by transient oxygen-glucose deprivation (OGD). Both 15 and 45 minutes of OGD were tolerated by control microRNA-expressing neurons but damaged >60% of neurons expressing SUMO2/3 microRNA. Damaging OGD (75 minutes) increased neuronal loss to 54% (control microRNA) and to 99% (SUMO2/3 microRNA). This suggests that activation of SUMO2/3 conjugation is an endogenous neuroprotective stress response.
Collapse
Affiliation(s)
- Anna Lena Datwyler
- Center for Stroke Research Berlin, Department of Experimental Neurology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | | | | | | | | | | | | | | |
Collapse
|
24
|
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
|
25
|
Zhang J, Freyer D, Rung O, Im AR, Hoffmann O, Dame C. Inflammation stimulates thrombopoietin (Tpo) expression in rat brain-derived microvascular endothelial cells, but suppresses Tpo in astrocytes and microglia. J Interferon Cytokine Res 2011; 30:465-9. [PMID: 20236022 DOI: 10.1089/jir.2009.0062] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Thrombopoietin (Tpo) and its receptor (c-Mpl; TpoR), which primary regulate megakaryopoiesis and platelet production, are also expressed in the central nervous system (CNS). Increased Tpo concentrations are present in the cerebrospinal fluid (CSF) of some patients with bacterial or viral meningitis. Since previous data implicated a proapoptotic role of Tpo on newly generated neuronal cells, we herein elucidated the regulation of Tpo in primary rat neurons (e17), astrocytes, and microglia (p0-p3), as well as in brain-derived vascular endothelial cells of 3-week-old rats after exposure to bacterial lipopolysaccharide (LPS). LPS inhibited Tpo gene expression in astrocytes and microglia, but not in neurons, most likely due to absence of Toll-like receptor 4 in neurons. While Tpo mRNA expression recovered in astrocytes after 24 h, it remained suppressed in microglia. Furthermore, we detected Tpo mRNA expression in primary brain-derived vascular endothelial cells, which also express the TpoR. In these cells, LPS significantly up-regulated Tpo mRNA expression. TpoR mRNA and protein expression remained constitutive in all cell types. Thus, our data provide evidence for a cell-type-specific modulation of Tpo mRNA expression by inflammation in brain-derived cells. Transient down-regulation of Tpo expression in astrocytes and microglia may limit Tpo-induced neuronal cell death in inflammatory brain disorders.
Collapse
Affiliation(s)
- Juan Zhang
- Department of Neonatology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | | | | | | | | | | |
Collapse
|
26
|
Li LZ, Bao YJ, Zhao M. 17beta-estradiol attenuates programmed cell death in cortical pericontusional zone following traumatic brain injury via upregulation of ERalpha and inhibition of caspase-3 activation. Neurochem Int 2010; 58:126-33. [PMID: 21093516 DOI: 10.1016/j.neuint.2010.11.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2010] [Revised: 11/04/2010] [Accepted: 11/09/2010] [Indexed: 01/01/2023]
Abstract
Pericontusional zone (PCZ) of traumatic cerebral contusion is a target of pharmacological intervention. It is well studied that 17beta-estradiol has a protective role in ischemic brain injury, but its role in brain protection of traumatic brain damage deserves further investigation, especially in pericontusional zone. Here we show that 17beta-estradiol enhances the protein expression and mRNA induction of estrogen alpha receptor (ERalpha) and prevents from programmed cell death in cortical pericontusional zone. ERalpha specific antagonist blocks this protective effect of 17beta-estradiol. Caspase-3 activation occurs in cortical pericontusional zone of the oil-treated injured rat brain and its activation is inhibited by 17beta-estradiol treatment. Additionally, ERalpha specific antagonist reverses this inhibition. Pan-caspase inhibitor also protect cortical pericontusional zone from programmed cell death. Our present study indicates 17beta-estradiol protects from programmed cell death in cortical pericontusional zone via enhancement of ERalpha and decrease of caspase-3 activation.
Collapse
Affiliation(s)
- Li-Zhuo Li
- Emergency Department, Shengjing Hospital, China Medical University, Shenyang, Liaoning, China.
| | | | | |
Collapse
|
27
|
Chu C, Xu B, Huang W. GnRH analogue attenuated apoptosis of rat hippocampal neuron after ischemia-reperfusion injury. J Mol Histol 2010; 41:387-93. [PMID: 20953819 DOI: 10.1007/s10735-010-9300-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2010] [Accepted: 10/05/2010] [Indexed: 12/12/2022]
Abstract
The expression and new functions of reproductive hormones in organs beyond hypothalamus-pituitary-gonad axis have been reported. So far, there is no report about the protective effects of GnRH analogue to hippocampal neurons suffering from ischemia-reperfusion injury. Middle cerebral artery occlusion model together with TUNEL staining were made in vivo and oxygen-glucose deprivation model together with double staining of Annexin V/PI with flow cytometer were made in vitro to observe the anti-apoptotic effects of GnRH analogue to hippocampal neurons after ischemia-reperfusion injury. The results found that the number of TUNEL positive pyramidal neurons in CA1 region in GnRH analogue experiment group was less than that in control group in vivo; the percentage of apoptotic neurons in GnRH analogue experiment group was less than that in control group in vitro. These findings suggested that pretreatment with certain concentration of GnRH analogue could attenuate apoptosis of hippocampal neurons. GnRH analogue has the protective effects to neurons.
Collapse
Affiliation(s)
- Chenyu Chu
- Department of Neurosurgery, Chinese PLA General Hospital, No. 28, FuXing Road, 100853 Beijing, China.
| | | | | |
Collapse
|
28
|
Abstract
Androgens can protect neurones from injury, although androgen neuroprotection is not well characterised in terms of either specificity or mechanism. In the present study, we compared the ability of androgens to protect neurones against a panel of insults, empirically determined to induce cell death by apoptotic or non-apoptotic mechanisms. Three criteria defining but not inclusive of apoptosis are: protection by caspase inhibition, protection by protein synthesis inhibition and the presence of pyknotic nuclei. According to these criteria, beta-amyloid, staurosporine, and Apoptosis Activator II induced cell death involving apoptosis, whereas hydrogen peroxide (H(2)O(2)), iron, calcium ionophore and 3-nitropropionic acid induced cell death featuring non-apoptotic characteristics. Pretreatment of hippocampal neurones with testosterone or dihydrotestosterone attenuated cell death induced by beta-amyloid, staurosporine and Apoptosis Activator II, but none of the other insults. The anti-oxidant Trolox did not reduce cell death induced by beta-amyloid, staurosporine and Apoptosis Activator II, but did protect against H(2)O(2) and iron. Similarly, a supra-physiological concentration of oestrogen reduced cell death induced by H(2)O(2) and iron, an effect not observed with androgens. We also show that activation of oestrogen pathways was not necessary for androgen neuroprotection. These data suggest that androgens directly activate a neuroprotective mechanism specific to inhibition of cell death involving apoptosis. Determining the specificity of androgen neuroprotection may enable the development of androgen compounds for the treatment of neurodegenerative disorders.
Collapse
Affiliation(s)
- T V Nguyen
- Neuroscience Graduate Programme and Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089-0191, USA
| | | | | | | |
Collapse
|
29
|
Gingerich S, Kim G, Chalmers J, Koletar M, Wang X, Wang Y, Belsham D. Estrogen receptor alpha and G-protein coupled receptor 30 mediate the neuroprotective effects of 17β-estradiol in novel murine hippocampal cell models. Neuroscience 2010; 170:54-66. [DOI: 10.1016/j.neuroscience.2010.06.076] [Citation(s) in RCA: 141] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2010] [Revised: 06/25/2010] [Accepted: 06/28/2010] [Indexed: 12/13/2022]
|
30
|
Ivanova A, Wuerfel J, Zhang J, Hoffmann O, Ballmaier M, Dame C. Expression pattern of the thrombopoietin receptor (Mpl) in the murine central nervous system. BMC DEVELOPMENTAL BIOLOGY 2010; 10:77. [PMID: 20667107 PMCID: PMC2921376 DOI: 10.1186/1471-213x-10-77] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2009] [Accepted: 07/28/2010] [Indexed: 11/10/2022]
Abstract
BACKGROUND Thrombopoietin (Thpo) and its receptor (Mpl), which regulate megakaryopoiesis, are expressed in the central nervous system (CNS), where Thpo is thought to exert pro-apoptotic effects on newly generated neurons. Mpl expression has been analysed in brain tissue on transcript level and in cultured primary rat neurons and astrocytes on protein level. Herein, we analysed Mpl expression in the developing and adult murine CNS by immunohistochemistry and investigated the brain of mice with homozygous Mpl deficiency (Mpl-/-) by MRI. RESULTS Mpl was not detectable at developmental stages E12 to E15 in any resident cells of the CNS. From E18 onwards, robust Mpl expression was found in various brain areas, including cerebral cortex, olfactory bulb, thalamus, hypothalamus, medulla, pons, and the grey matter of spinal cord. However, major developmental changes became obvious: In the subventricular zone of the cerebral cortex Mpl expression occurred only during late gestation, while in the hippocampus Mpl expression was detectable for first time at stage P4. In the white matter of the cerebellum Mpl expression was restricted to the perinatal period. In the adult cerebellum, Mpl expression switched to Purkinje cell. The majority of other Mpl-positive cells were NeuN-positive neurons. None of the cells could be double-labelled with astrocyte marker GFAP. Mpl-/- mice showed no gross abnormalities of the brain. CONCLUSIONS Our data locate Mpl expression to neurons at different subdivisions of the spinal cord, rhombencephalon, midbrain and prosencephalon. Besides neuronal cells Mpl protein is also expressed in Purkinje cells of the adult cerebellum.
Collapse
Affiliation(s)
- Anna Ivanova
- Department of Neonatology, Charité - Universitätsmedizin, Germany
| | | | | | | | | | | |
Collapse
|
31
|
Alcohol withdrawal and brain injuries: beyond classical mechanisms. Molecules 2010; 15:4984-5011. [PMID: 20657404 PMCID: PMC6257660 DOI: 10.3390/molecules15074984] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2010] [Revised: 07/15/2010] [Accepted: 07/19/2010] [Indexed: 01/12/2023] Open
Abstract
Unmanaged sudden withdrawal from the excessive consumption of alcohol (ethanol) adversely alters neuronal integrity in vulnerable brain regions such as the cerebellum, hippocampus, or cortex. In addition to well known hyperexcitatory neurotransmissions, ethanol withdrawal (EW) provokes the intense generation of reactive oxygen species (ROS) and the activation of stress-responding protein kinases, which are the focus of this review article. EW also inflicts mitochondrial membranes/membrane potential, perturbs redox balance, and suppresses mitochondrial enzymes, all of which impair a fundamental function of mitochondria. Moreover, EW acts as an age-provoking stressor. The vulnerable age to EW stress is not necessarily the oldest age and varies depending upon the target molecule of EW. A major female sex steroid, 17β-estradiol (E2), interferes with the EW-induced alteration of oxidative signaling pathways and thereby protects neurons, mitochondria, and behaviors. The current review attempts to provide integrated information at the levels of oxidative signaling mechanisms by which EW provokes brain injuries and E2 protects against it.
Collapse
|
32
|
Picazo O, Becerril-Montes A, Huidobro-Perez D, Garcia-Segura LM. Neuroprotective actions of the synthetic estrogen 17alpha-ethynylestradiol in the hippocampus. Cell Mol Neurobiol 2010; 30:675-82. [PMID: 20044777 PMCID: PMC11498788 DOI: 10.1007/s10571-009-9490-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2009] [Accepted: 12/15/2009] [Indexed: 12/23/2022]
Abstract
17alpha-ethynylestradiol (EE2), a major constituent of many oral contraceptives, is similar in structure to 17beta-estradiol, which has neuroprotective properties in several animal models. This study explored the potential neuroprotective actions of EE2 against kainic and quinolinic acid toxicity in the hippocampus of adult ovariectomized Wistar rats. A decrease in the number of Nissl-stained neurons and the induction of vimentin immunoreactivity in astrocytes was observed in the hilus of the dentate gyrus of the hippocampus after the administration of either kainic acid or quinolinic acid. EE2 prevented the neuronal loss and the induction of vimentin immunoreactivity induced by kainic acid at low (1 microg/rat) and high (10-100 microg/rat) doses and exerted a protection against quinolinic acid toxicity at a low dose (1 microg/rat) only. These observations demonstrate that EE2 exerts neuroprotective actions against excitotoxic insults. This finding is relevant for the design of new neuroprotective estrogenic compounds.
Collapse
Affiliation(s)
- Ofir Picazo
- Escuela Superior de Medicina del Instituto Politécnico Nacional, Plan de San Luis y Diaz Mirón Col. Sto. Tomás, 11340 Mexico, DF Mexico
| | - Adriana Becerril-Montes
- Escuela Superior de Medicina del Instituto Politécnico Nacional, Plan de San Luis y Diaz Mirón Col. Sto. Tomás, 11340 Mexico, DF Mexico
| | - Delia Huidobro-Perez
- Escuela Superior de Medicina del Instituto Politécnico Nacional, Plan de San Luis y Diaz Mirón Col. Sto. Tomás, 11340 Mexico, DF Mexico
| | | |
Collapse
|
33
|
Manthey D, Gamerdinger M, Behl C. The selective beta1-adrenoceptor antagonist nebivolol is a potential oestrogen receptor agonist with neuroprotective abilities. Br J Pharmacol 2010; 159:1264-73. [PMID: 20128815 DOI: 10.1111/j.1476-5381.2009.00610.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND AND PURPOSE Nebivolol, a selective beta(1)-adrenoceptor antagonist mediating rapid vasodilating effects, is used clinically to treat hypertension. Recently, it was reported that nebivolol also acts as an oestrogen receptor (ER) agonist. To investigate the neuroprotective potential of oestrogens, we assessed the oestrogenic effects of nebivolol in several in vitro neuronal models. EXPERIMENTAL APPROACH Human neuroepithelioma SK-N-MC cells stably transfected with human ER alpha and beta, and mouse N2A neuroblastoma cells expressing human APP695(SWE)[N2Aswe, stably transfected with the Swedish mutation form of the Alzheimer-associated amyloid precursor protein (APPswe, K670M/N671L)] were incubated with different concentrations of nebivolol and 17beta-oestradiol (E2) for 24-48 h. ER activation was detected in a specific reporter assay, and ER-dependent gene expression was measured by quantitative real-time PCR (qRT PCR). Furthermore, cell survival rates were determined, and oxidative stress was induced by hydrogen peroxide and paraquat. Amyloid beta protein precursor (APP) processing was investigated, and the cleavage fragments sAPPalpha and Abeta were quantified via alpha-, beta- and gamma-secretase activity assays. Alterations of secretase expression levels were determined by qRT PCR. KEY RESULTS Nebivolol induces oestrogen-dependent gene transcription, and protects neuronal cells against oxidative stress even at low and physiological concentrations (10(-8) M). Moreover, nebivolol modulates processing of APP in mouse neuronal N2Aswe cells by increasing alpha-secretase activity, ultimately leading to enhanced release of soluble non-amyloidogenic sAPPalpha. CONCLUSIONS AND IMPLICATIONS We showed that nebivolol acts as ER agonist in neuronal cell lines, and suggest oestrogen-like neuroprotective effects mediated by nebivolol.
Collapse
Affiliation(s)
- D Manthey
- Department of Pathobiochemistry, University Medical Center, Johannes Gutenberg-University, Mainz, Germany
| | | | | |
Collapse
|
34
|
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
|
35
|
Alvarez-Delgado C, Mendoza-Rodríguez CA, Picazo O, Cerbón M. Different expression of alpha and beta mitochondrial estrogen receptors in the aging rat brain: interaction with respiratory complex V. Exp Gerontol 2010; 45:580-5. [PMID: 20096765 DOI: 10.1016/j.exger.2010.01.015] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2009] [Revised: 01/11/2010] [Accepted: 01/14/2010] [Indexed: 01/11/2023]
Abstract
Recent evidence suggests that hormonal effects on mitochondria could be mediated by mitochondrial estrogen receptors (mtERs). These receptors are new candidates for the beneficial estrogenic effects on mitochondria in different physiological conditions. The aim of this investigation was to study mtER expression during brain aging. We analyzed mtERalpha and mtERbeta expression in cortical, hippocampal and hypothalamic mitochondria of young adult (3months) and aged (18 months) female Wistar rats by Western blot. In addition, we explored the interaction of mtERbeta with respiratory complex V by using coimmunoprecipitation assays. The results show that mtERalpha and mtERbeta are present in young and aged brain mitochondria. We also demonstrate that mtERs are expressed as variants and have a brain region specific distribution. The predominant mtER variants detected were of 61 and 55KDa for mtERalpha and of 63 and 52KDa for mtERbeta. However, we did not observe differences in the mtERalpha or beta content between the two age groups studied. Additionally, we show that mtERbeta interacts with complex V. The overall results demonstrate that there is a differential expression of mtERalpha and mtERbeta variants in different brain areas, indicating that they may participate in different functions in the brain during aging.
Collapse
Affiliation(s)
- Carolina Alvarez-Delgado
- Facultad de Química, Departamento de Biología, Universidad Nacional Autónoma de México, México D.F., Mexico
| | | | | | | |
Collapse
|
36
|
Gold SM, Voskuhl RR. Estrogen treatment in multiple sclerosis. J Neurol Sci 2009; 286:99-103. [PMID: 19539954 PMCID: PMC2760629 DOI: 10.1016/j.jns.2009.05.028] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2009] [Revised: 05/11/2009] [Accepted: 05/19/2009] [Indexed: 10/20/2022]
Abstract
Currently available treatments for multiple sclerosis (MS) reduce inflammatory lesions on MRI and decrease clinical relapses but have limited effects on disability. Novel treatment options that target both the inflammatory as well as the neurodegenerative component of the disease are therefore needed. A growing body of evidence from basic science and clinical studies supports the therapeutic potential of estrogens in MS. Mechanisms of action include both immunomodulatory and directly neuroprotective pathways. A first pilot trial of oral estriol treatment showed encouraging results. There are now several phase II trials underway to further determine the efficacy of estrogen treatment in MS.
Collapse
Affiliation(s)
- Stefan M Gold
- Multiple Sclerosis Program, Department of Neurology, and Cousins Center, Geffen School of Medicine, University of California Los Angeles, U.S.A
| | - Rhonda R Voskuhl
- Multiple Sclerosis Program, Department of Neurology, and Cousins Center, Geffen School of Medicine, University of California Los Angeles, U.S.A
| |
Collapse
|
37
|
Effects of estrogen on AF64A-induced apoptosis in NG108-15 cells. Brain Res 2009; 1297:9-16. [PMID: 19729002 DOI: 10.1016/j.brainres.2009.08.065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2009] [Revised: 08/18/2009] [Accepted: 08/24/2009] [Indexed: 11/23/2022]
Abstract
In this study, we show that pretreatment with physiological concentrations (1-100 nM) of 17beta-estradiol decreased apoptosis induced by ethylcholine aziridinium (AF64A), a choline toxin, in the cholinergic neuronal cell line NG108-15. These protective effects were observed after short-term (30 min) pretreatment, and were blocked by treatment with an estrogen receptor antagonist and inhibitors of phosphatidylinositol 3-kinase (PI3K) and mitogen-activated protein kinase kinase (MEK). The protective effects were, however, not reversed by a protein synthesis inhibitor. Furthermore, we examined the effects of 17beta-estradiol on choline uptake in NG108-15 cells. Although choline uptake was inhibited by a selective inhibitor of choline uptake, hemicholinium-3, it was not altered by treatment with 17beta-estradiol. These results indicated that the protective effect of 17beta-estradiol on AF64A-induced apoptosis could be nongenomic, and that this effect may be due to the activation of PI3K/Akt and/or MEK/extracellular signal-regulated kinase (ERK) pathways.
Collapse
|
38
|
Orellano EA, Rivera OJ, Chevres M, Chorna NE, González FA. Inhibition of neuronal cell death after retinoic acid-induced down-regulation of P2X7 nucleotide receptor expression. Mol Cell Biochem 2009; 337:83-99. [PMID: 19882109 DOI: 10.1007/s11010-009-0288-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2009] [Accepted: 10/08/2009] [Indexed: 01/12/2023]
Abstract
Apoptosis is a major mechanism for cell death in the nervous system during development. P2X(7) nucleotide receptors are ionotropic ATP receptors that mediate cell death under pathological conditions. We developed an in vitro protocol to investigate the expression and functional responses of P2X(7) nucleotide receptors during retinoic acid (RA)-induced neuronal differentiation of human SH-SY5Y neuroblastoma cells. Neuronal differentiation was examined measuring cellular growth arrest and neuritic processes elongation. We found that SH-SY5Y cells treated for 5 days with RA under low serum content exhibited a neuron-like phenotype with neurites extending more than twice the length of the cell body and cell growth arrest. Concurrently, we detected the abolishment of intracellular-free calcium mobilization and the down-regulation of P2X(7) nucleotide receptor protein expression that protected differentiated cells from neuronal cell death and reduced caspase-3 cleavage-induced by P2X(7) nucleotide receptor agonist. The role of P2X(7) nucleotide receptors in neuronal death was established by selectively antagonizing the receptor with KN-62 prior to its activation. We assessed the involvement of protein kinases and found that p38 signaling was activated in undifferentiated after nucleotide stimulation, but abolished by the differentiating RA pretreatment. Importantly, P2X(7) receptor-induced caspase-3 cleavage was blocked by the p38 protein kinase specific inhibitor PD169316. Taken together, our results suggest that RA treatment of human SH-SY5Y cells leads to decreased P2X(7) nucleotide receptor protein expression thus protecting differentiated cells from extracellular nucleotide-induced neuronal death, and p38 signaling pathway is critically involved in this protection of RA-differentiated cells.
Collapse
Affiliation(s)
- Elsie A Orellano
- Department of Biology, University of Puerto Rico, Río Piedras Campus, San Juan, PR, USA
| | | | | | | | | |
Collapse
|
39
|
Lorenz L, Dang J, Misiak M, Tameh Abolfazl A, Beyer C, Kipp M. Combined 17beta-oestradiol and progesterone treatment prevents neuronal cell injury in cortical but not midbrain neurones or neuroblastoma cells. J Neuroendocrinol 2009; 21:841-9. [PMID: 19686448 DOI: 10.1111/j.1365-2826.2009.01903.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Oestrogens are powerful endogenous and exogenous neuroprotective hormones in animal models of brain injury, including focal cerebral ischaemia. This protective effect has been demonstrated under a variety of different treatments and injury paradigms, such as in vivo and in vitro stroke conditions. Neuroprotection in the central nervous system by progesterone is less defined. In the present study, cultured cortical and midbrain mouse neurones and human neuroblastoma cells (SH-SY5Y) were exposed to combined glucose-serum deprivation (CGSD), which is regarded as a reliable model mimicking the effects of ischaemia in vitro. Cell viability was assayed using lactate dehydrogenase release and metabolic activity. Conditions for CGSD treatment were chosen to yield half-maximal cell death rates. The validity of CGSD in vitro was compared with permanent middle cerebral artery occlusion (MCAO) in vivo. CGSD for 4 h induced half-maximal neuronal cell death. MCAO in vivo for the same period resulted in significant neuronal loss, also suggesting the validity of CGSD as a suitable stroke-like in vitro model. Combined steroid treatment (17beta-oestradiol and progesterone) but not the application of single steroids abolished CGSD-induced cell death of cortical neurones in vitro. By contrast, no cell protection was found in midbrain neurones or neuroblastoma cells. The co-application of oestrogen (ICI 182,780) or progesterone (RU-486) receptor antagonists did not obviously counteract the protective steroid effects. This suggests the operation of nonclassical steroid mechanisms and their implication in mediation of hormonal effects. The surplus of combined protective hormonal effects might be a result of the observed influence of progesterone application on neuronal oestradiol synthesis. The data obtained in the present study clearly highlight the potential of a combined steroid treatment under toxic degenerative brain pathologies.
Collapse
Affiliation(s)
- L Lorenz
- Institute of Neuroanatomy, Faculty of Medicine, RWTH Aachen University, Aachen, Germany.
| | | | | | | | | | | |
Collapse
|
40
|
Pastural E, Ritchie S, Lu Y, Jin W, Kavianpour A, Khine Su-Myat K, Heath D, Wood PL, Fisk M, Goodenowe DB. Novel plasma phospholipid biomarkers of autism: mitochondrial dysfunction as a putative causative mechanism. Prostaglandins Leukot Essent Fatty Acids 2009; 81:253-64. [PMID: 19608392 DOI: 10.1016/j.plefa.2009.06.003] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2009] [Revised: 06/12/2009] [Accepted: 06/15/2009] [Indexed: 12/30/2022]
Abstract
Autism is a neurological disorder that manifests as noticeable behavioral and developmental abnormalities predominantly in males between the ages of 2 and 10. Although the genetics, biochemistry and neuropathology of this disease have been extensively studied, underlying causal factors to this disease have remained elusive. Using a longitudinal trial design in which three plasma samples were collected from 15 autistic and 12 non-autistic age-matched controls over the course of 1 year, universal and unambiguous alterations in lipid metabolism were observed. Biomarkers of fatty acid elongation and desaturation (poly-unsaturated long chain fatty acids (PUFA) and/or saturated very long chain fatty acids (VLCFA)-containing ethanolamine phospholipids) were statistically elevated in all autistic subjects. In all 8 of the affected/non-affected sibling pairs, the affected sibling had higher levels of these biomarkers than the unaffected sibling. Exposure of neurons, astrocytes and hepatocytes in vitro to elevated extracellular glutamate levels resulted in lipid biomarker changes indistinguishable from those observed in autistic subjects. Glutamate stress also resulted in in vitro decreased levels of reduced glutathione (GSH), methionine and cysteine, in a similar way to the decreases we observed in autism plasma. Impaired mitochondrial fatty acid oxidation, elevated plasma VLCFAs, and glutamate toxicity as putative causal factors in the biochemistry, neuropathology, and gender bias in autism are discussed.
Collapse
Affiliation(s)
- Elodie Pastural
- Phenomenome Discoveries Inc., 204-407 Downey Road, Saskatoon, Saskatchewan, Canada S7N 4L8
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Bethea CL, Reddy AP, Tokuyama Y, Henderson JA, Lima FB. Protective actions of ovarian hormones in the serotonin system of macaques. Front Neuroendocrinol 2009; 30:212-38. [PMID: 19394356 PMCID: PMC2704571 DOI: 10.1016/j.yfrne.2009.04.003] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2009] [Revised: 04/14/2009] [Accepted: 04/15/2009] [Indexed: 12/19/2022]
Abstract
The serotonin neurons of the dorsal and medial raphe nuclei project to all areas of the forebrain and play a key role in mood disorders. Hence, any loss or degeneration of serotonin neurons could have profound ramifications. In a monkey model of surgical menopause with hormone replacement and no neural injury, E and P decreased gene expression in the dorsal raphe nucleus of c-jun n-terminal kinase (JNK1) and kynurenine mono-oxygenase (KMO) that promote cell death. In concert, E and P increased gene expression of superoxide dismutase (SOD1), VEGF, and caspase inhibitory proteins that promote cellular resilience in the dorsal raphe nucleus. Subsequently, we showed that ovarian steroids inhibit pivotal genes in the caspase-dependent and caspase-independent pathways in laser-captured serotonin neurons including apoptosis activating factor (Apaf1), apoptosis-inducing factor (AIF) and second mitochondria-derived activator of caspases (Smac/Diablo). SOD1 was also increased specifically in laser-captured serotonin neurons. Examination of protein expression in the dorsal raphe block revealed that JNK1, phosphoJNK1, AIF and the translocation of AIF from the mitochondria to the nucleus decreased with hormone therapy, whereas pivotal execution proteins in the caspase pathway were unchanged. In addition, cyclins A, B, D1 and E were inhibited, which would prevent re-entry into the cell cycle and catastrophic death. These data indicated that in the absence of gross injury to the midbrain, ovarian steroids inhibit the caspase-independent pathway and cell cycle initiation in serotonin neurons. To determine if these molecular actions prevented cellular vulnerability or death, we examined DNA fragmentation in the dorsal raphe nucleus with the TUNEL assay (terminal deoxynucleotidyl transferase nick end labeling). Ovarian steroids significantly decreased the number of TUNEL-positive cells in the dorsal raphe. Moreover, TUNEL staining prominently colocalized with TPH immunostaining, a marker for serotonin neurons. In summary, ovarian steroids increase the cellular resilience of serotonin neurons and may prevent serotonin neuron death in women facing decades of life after menopause. The survival of serotonin neurons would support cognition and mental health.
Collapse
Affiliation(s)
- Cynthia L Bethea
- Divisions of Reproductive Sciences, Oregon National Primate Research Center, Beaverton, OR 97006, United States.
| | | | | | | | | |
Collapse
|
42
|
Sribnick EA, Del Re AM, Ray SK, Woodward JJ, Banik NL. Estrogen attenuates glutamate-induced cell death by inhibiting Ca2+ influx through L-type voltage-gated Ca2+ channels. Brain Res 2009; 1276:159-70. [PMID: 19389388 PMCID: PMC2700344 DOI: 10.1016/j.brainres.2009.04.022] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2009] [Revised: 04/02/2009] [Accepted: 04/04/2009] [Indexed: 12/26/2022]
Abstract
Estrogen-mediated neuroprotection is observed in neurodegenerative disease and neurotrauma models; however, determining a mechanism for these effects has been difficult. We propose that estrogen may limit cell death in the nervous system tissue by inhibiting increases in intracellular free Ca(2+). Here, we present data using VSC 4.1 cell line, a ventral spinal motoneuron and neuroblastoma hybrid cell line. Treatment with 1 mM glutamate for 24 h induced apoptosis. When cells were pre-treated with 100 nM 17beta-estradiol (estrogen) for 1 h and then co-treated with glutamate, apoptotic death was significantly attenuated. Estrogen also prevented glutamate-mediated changes in resting membrane potential and membrane capacitance. Treatment with either 17 alpha-estradiol or cell impermeable estrogen did not mimic the findings seen with estrogen. Glutamate treatment significantly increased both intracellular free Ca(2+) and the activities of downstream proteases such as calpain and caspase-3. Estrogen attenuated both the increases in intracellular free Ca(2+) and protease activities. In order to determine the pathway responsible for estrogen-mediated inhibition of these increases in intracellular free Ca(2+), cells were treated with several Ca(2+) entry inhibitors, but only the L-type Ca(2+) channel blocker nifedipine demonstrated cytoprotective effects comparable to estrogen. To expand these findings, cells were treated with the L-type Ca(2+) channel agonist FPL 64176, which increased both cell death and intracellular free Ca(2+), and estrogen inhibited both effects. From these observations, we conclude that estrogen limits glutamate-induced cell death in VSC 4.1 cells through effects on L-type Ca(2+) channels, inhibiting Ca(2+) influx as well as activation of the pro-apoptotic proteases calpain and caspase-3.
Collapse
Affiliation(s)
- Eric A. Sribnick
- Division of Neurology, Department of Neurosciences, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Angelo M. Del Re
- Division of Neurology, Department of Neurosciences, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Swapan K. Ray
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC 29209, USA
| | - John J. Woodward
- Division of Neurology, Department of Neurosciences, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Naren L. Banik
- Division of Neurology, Department of Neurosciences, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
43
|
Xia Y, Xing JZ, Krukoff TL. Neuroprotective effects of R,R-tetrahydrochrysene against glutamate-induced cell death through anti-excitotoxic and antioxidant actions involving estrogen receptor-dependent and -independent pathways. Neuroscience 2009; 162:292-306. [PMID: 19410635 DOI: 10.1016/j.neuroscience.2009.04.068] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2009] [Revised: 04/21/2009] [Accepted: 04/28/2009] [Indexed: 01/21/2023]
Abstract
Glutamate-induced neural cell death is mediated by excitotoxicity and oxidative stress. Treatment of glutamate toxicity with estrogen and its related compounds for neuroprotection remains controversial. In this study, we examined the effects of selective estrogen receptor (ER) ligands on glutamate toxicity and found that R,R-tetrahydrochrysene (R,R-THC), an antagonist of ERbeta and agonist of ERalpha, has neuroprotective effects against glutamate-induced death in primary rat cortical cells and mouse N29/4 hypothalamic cells. The protective effect of R,R-THC was dose-dependent and was maintained even when added several hours after the initial glutamate exposure. R,R-THC blocked glutamate-induced depletion of intracellular glutathione, increased superoxide dismutase activity, and protected cells from hydrogen peroxide-induced death. R,R-THC also prevented glutamate-induced nuclear translocation of apoptotic inducing factor and release of mitochondrial cytochrome c. The protective effect of R,R-THC was blocked by methyl-piperidino-pyrazole (MPP; an ERalpha antagonist) in glutamate-treated cortical cells, and pretreatment with MK-801 (an NMDA receptor antagonist) but not CNQX (an AMPA/kainate receptor antagonist) increased cell survival. On the other hand, MPP did not block the protective effect of R,R-THC in glutamate-treated N29/4 cells, and neither MK-801 nor CNQX conferred protection. Activation of ERalpha and/or ERbeta with 17beta-estradiol (E2), propyl-pyrazole-triol or diarylpropionitrile did not provide effective neuroprotection, and pretreatment with ICI 182,780 did not inhibit the protective effect of R,R-THC in either type of cell. These results suggest that the use of ER agonists (including E2) has limited beneficial effects when both excitotoxicity and oxidative stress occur. In contrast to agonists of ERs, R,R-THC, which possesses anti-excitotoxic and antioxidant actions via ER-dependent and -independent pathways, provides significant neuroprotection.
Collapse
Affiliation(s)
- Y Xia
- Department of Cell Biology and Center for Neuroscience, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada T6G 2H7.
| | | | | |
Collapse
|
44
|
Bebo BF, Dehghani B, Foster S, Kurniawan A, Lopez FJ, Sherman LS. Treatment with selective estrogen receptor modulators regulates myelin specific T-cells and suppresses experimental autoimmune encephalomyelitis. Glia 2009; 57:777-90. [PMID: 19031437 PMCID: PMC2667209 DOI: 10.1002/glia.20805] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Steroidal estrogens can regulate inflammatory immune responses and may be involved in the suppression of multiple sclerosis (MS) during pregnancy. However, the risks and side effects associated with steroidal estrogens may limit their usefulness for long-term MS therapy. Selective estrogen receptor modulators (SERMs) could provide an alternative therapeutic strategy, because they behave as estrogen agonists in some tissues, but are either inert or behave like estrogen antagonists in other tissues. In this study, we investigated the ability of two commercially available SERMs (tamoxifen and raloxifene) to regulate myelin specific immunity and experimental autoimmune encephalomyelitis (EAE) in mice. Both tamoxifen and raloxifene suppressed myelin antigen specific T-cell proliferation. However, tamoxifen was more effective in this regard. Tamoxifen treatment reduced the induction of major histocompatibility complex II by lipopolysaccharide stimulated dendritic cells and decreased their ability to activate myelin specific T-cells. At lower doses, tamoxifen was found to increase the levels of Th2 transcription factors and induce a Th2 bias in cultures of myelin-specific splenocytes. EAE symptoms and the degree of demyelination were less severe in mice treated with tamoxifen than in control mice. These findings support the notion that tamoxifen or related SERMs are potential agents that could be used in the treatment of inflammatory autoimmune disorders that affect the central nervous system.
Collapse
Affiliation(s)
- Bruce F. Bebo
- Neurological Sciences Institute, Oregon Health & Science University, Beaverton, OR
- Department of Neurology, Oregon Health & Science University, Portland, OR
| | - Babak Dehghani
- Neurological Sciences Institute, Oregon Health & Science University, Beaverton, OR
| | - Scott Foster
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR
| | - Astrid Kurniawan
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR
| | | | - Larry S. Sherman
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR
- Department of Cell and Developmental Biology, Oregon Health & Science University, Portland, OR
| |
Collapse
|
45
|
Schreihofer DA, Redmond L. Soy phytoestrogens are neuroprotective against stroke-like injury in vitro. Neuroscience 2009; 158:602-9. [PMID: 18976694 PMCID: PMC2652887 DOI: 10.1016/j.neuroscience.2008.10.003] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2008] [Revised: 09/30/2008] [Accepted: 10/06/2008] [Indexed: 11/19/2022]
Abstract
Diets high in soy are neuroprotective in experimental stroke. This protective effect is hypothesized to be mediated by phytoestrogens contained in soy, because some of these compounds have neuroprotective effects in in vitro models of cell death. We tested the ability of the soy phytoestrogens genistein, daidzein, and the daidzein metabolite equol to protect embryonic rat primary cortical neurons from ischemic-like injury in vitro at doses typical of circulating concentrations in human populations (0.1-1 microM). All three phytoestrogens inhibited lactate dehydrogenase (LDH) release from cells exposed to glutamate toxicity or the calcium-ATPase inhibitor, thapsigargin. In cells exposed to hypoxia or oxygen-glucose deprivation (OGD), pretreatment with the phytoestrogens inhibited cell death in an estrogen receptor (ER) dependent manner. Although OGD results in multiple modes of cell death, examination of alpha-spectrin cleavage and caspase-3 activation revealed that the phytoestrogens were able to inhibit apoptotic cell death in this model. In addition, blockade of phosphoinositide 3-kinase prevented the protective effects of genistein and daidzein, and blockade of mitogen-activated protein kinase prevented genistein-dependent neuroprotection. These results suggest that pretreatment with dietary levels of soy phytoestrogens can mimic neuroprotective effects observed with estrogen and appear to use the same ER-kinase pathways to inhibit apoptotic cell death.
Collapse
Affiliation(s)
- D A Schreihofer
- Department of Physiology, CA3145, Medical College of Georgia, 1120 15th Street, Augusta, GA 30912-3000, USA.
| | | |
Collapse
|
46
|
Aryl hydrocarbon receptor-mediated apoptosis of neuronal cells: A possible interaction with estrogen receptor signaling. Neuroscience 2009; 158:811-22. [DOI: 10.1016/j.neuroscience.2008.10.045] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2008] [Revised: 10/27/2008] [Accepted: 10/30/2008] [Indexed: 01/21/2023]
|
47
|
Abstract
It has been known for decades that females are more susceptible than men to inflammatory autoimmune diseases, including multiple sclerosis (MS), rheumatoid arthritis, and psoriasis. In addition, female patients with these diseases experience clinical improvements during pregnancy with a temporary "rebound" exacerbation postpartum. These clinical observations indicate an effect of sex hormones on disease and suggest the potential use of the male hormone testosterone and the pregnancy hormone estriol, respectively, for the treatment of MS. A growing number of studies using the MS animal model experimental autoimmune encephalomyelitis (EAE) support a therapeutic effect of these hormones. Both testosterone and estriol have been found to induce anti-inflammatory as well as neuroprotective effects. Findings from two recent pilot studies of transdermal testosterone in male MS patients and oral estriol in female MS patients are encouraging. In this paper, we review the preclinical and clinical evidence for sex hormone treatments in MS and discuss potential mechanisms of action.
Collapse
Affiliation(s)
- Stefan M Gold
- Multiple Sclerosis Program, Department of Neurology, and Cousins Center, Geffen School of Medicine, University of California Los Angeles, Neurosci Res Bldg 1, 4 Floor, 635 Charles E Young Dr S, Los Angeles, CA 90095, U.S.A
| | - Rhonda R Voskuhl
- Multiple Sclerosis Program, Department of Neurology, and Cousins Center, Geffen School of Medicine, University of California Los Angeles, Neurosci Res Bldg 1, 4 Floor, 635 Charles E Young Dr S, Los Angeles, CA 90095, U.S.A
| |
Collapse
|
48
|
Patchev VK, Bachurin SO, Albers M, Fritzemeier KH, Papadopoulos V. Neurotrophic estrogens: essential profile and endpoints for drug discovery☆. Drug Discov Today 2008; 13:734-47. [DOI: 10.1016/j.drudis.2008.03.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2008] [Revised: 03/04/2008] [Accepted: 03/12/2008] [Indexed: 11/15/2022]
|
49
|
Foster TC, Rani A, Kumar A, Cui L, Semple-Rowland SL. Viral vector-mediated delivery of estrogen receptor-alpha to the hippocampus improves spatial learning in estrogen receptor-alpha knockout mice. Mol Ther 2008; 16:1587-93. [PMID: 18594506 PMCID: PMC2638757 DOI: 10.1038/mt.2008.140] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2007] [Accepted: 03/06/2008] [Indexed: 01/01/2023] Open
Abstract
Estrogen, which influences both classical genomic and rapid membrane-associated signaling cascades, has been implicated in the regulation of hippocampal function, including spatial learning. Gene mutation studies suggest that estrogen effects are mediated by estrogen receptor-alpha (ER-alpha); however, because gonadal steroids influence the organization of the hippocampus during development, it has been difficult to distinguish developmental effects from those specific to adults. In this study we show that lentiviral delivery of the gene encoding ER-alpha to the hippocampus of adult ER-alpha-knockout (ER-alphaKO) mice restores hippocampal responsiveness to estrogen and rescues spatial learning. We propose that constitutive estrogen receptor activity is important for maintaining hippocampus-dependent memory function in adults.
Collapse
Affiliation(s)
- Thomas C Foster
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, Florida 32610-0244, USA.
| | | | | | | | | |
Collapse
|
50
|
Araújo GW, Beyer C, Arnold S. Oestrogen influences on mitochondrial gene expression and respiratory chain activity in cortical and mesencephalic astrocytes. J Neuroendocrinol 2008; 20:930-41. [PMID: 18445124 DOI: 10.1111/j.1365-2826.2008.01747.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The regulation of mitochondrial energy metabolism plays an essential role in the central nervous system (CNS). Abnormalities of the mitochondrial respiratory chain often accompany neurodegenerative diseases. This makes mitochondria a perfect target for strategies of cellular protection against toxic compounds and pathological conditions. Steroid hormones, such as oestrogen, are well-known to fulfil a protective role in the brain during ischaemic and degenerative processes. Because astrocytes function as the major energy supplier in the CNS, we have analysed oestrogen effects on the mitochondrial respiratory chain of this cell type. In our studies, we applied semi- and quantitative polymerase chain reaction analysis of gene expression and polarographic measurements of the respiratory chain activity of mitochondria. We observed that structural and functional properties were regulated dependent on the oestrogen exposure time and the brain region, but independent of the nuclear oestrogen receptors. We could demonstrate that long-term oestrogen exposure increases the subunit gene expression of respiratory chain complexes and the mitochondrial DNA content, thereby indicating an up-regulation of the amount of mitochondria per cell together with an increase of mitochondrial energy production. This could represent an important indirect mechanism by which long-term oestrogen exposure protects neurones from cell death under neurotoxic conditions. On the other hand, we observed short-term effects of oestrogen on the activity of mitochondrial, proton-pumping respiratory chain complexes. In astrocytes from the cortex, respiratory chain activity was decreased, whereas it was increased in astrocytes from the mesencephalon. An increased production of reactive oxygen species would be the consequence of an increased respiratory chain activity in mesencephalic astrocytes. This could explain the different efficiencies of oestrogen-mediated short-term protection in distinct brain regions, but also indicates the limitations for a therapeutic short-term application of oestrogen.
Collapse
Affiliation(s)
- G W Araújo
- Institute for Neuroanatomy, Faculty of Medicine, RWTH, Aachen, Germany
| | | | | |
Collapse
|