1
|
Vázquez-Sola A, Torres-Torrelo H, Yagüe JG. Membrane progesterone and oestrogen receptors modulate GABAergic transmission in the prefrontal cortex of prepubertal male, but not female, mice. Exp Physiol 2025. [PMID: 40309895 DOI: 10.1113/ep092439] [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: 11/14/2024] [Accepted: 12/18/2024] [Indexed: 05/02/2025]
Abstract
The sex hormones progesterone (P) and oestrogen (E) reorganize GABAergic transmission in the prefrontal cortex (PFC) during the transition from childhood to adolescence, generating a new excitatory-inhibitory balance necessary for the computational capacity of the mature PFC. Little is known, however, about the hormone receptors involved or whether there are sex differences in the modulation of GABAergic transmission they exert. We hypothesize that P and E can rapidly (within minutes) modulate GABAergic currents through G protein-coupled receptors, namely membrane P receptors (mPRs) and the G protein-coupled E receptor (GPER), respectively, in PFC. First, we quantified the expression of P and E receptors in PFC using quantitative RT-PCR. Secondly, we recorded synaptic (phasic) and extrasynaptic (tonic) GABAergic currents in basal conditions and in response to the activation of mPRs and GPER using patch-clamp recordings in PFC neurons of prepubertal female and male mice. Expression levels of mPRs differed in the PFC of females and males, but no differences were found in the basal levels of phasic or tonic GABAergic currents between sexes. Interestingly, selective activation of mPRs increased tonic GABAergic transmission in males but not in females, and activation of GPER increased phasic GABAergic transmission only in males. We also demonstrated that GABAergic modulation exerted by mPRs and GPER was dependent on protein kinase A and C. This study sheds light on new mechanisms by which P and E can rapidly modulate GABAergic transmission in PFC neurons through the activation of mPRs and GPER.
Collapse
Affiliation(s)
- Aitana Vázquez-Sola
- Grupo de Neurofisiología Celular, Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe s/n, Madrid, Spain
| | - Hortensia Torres-Torrelo
- Grupo de Neurofisiología Celular, Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe s/n, Madrid, Spain
| | - Josué García Yagüe
- Grupo de Neurofisiología Celular, Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe s/n, Madrid, Spain
| |
Collapse
|
2
|
Zhang X, Mao J, Li H, Zhang C, Ge H, Zhong J. Activation of GPER1 alleviates white matter injury by promoting microglia M2 polarization through EGFR/Stat3 pathway in intracerebral hemorrhage mice. J Stroke Cerebrovasc Dis 2025; 34:108315. [PMID: 40228567 DOI: 10.1016/j.jstrokecerebrovasdis.2025.108315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2025] [Revised: 04/09/2025] [Accepted: 04/11/2025] [Indexed: 04/16/2025] Open
Abstract
BACKGROUND White matter injury (WMI) is a major pathophysiological process after intracerebral hemorrhage (ICH). G protein-coupled estrogen receptor 1 (GPER1) has been validated to exert a crucial role in regulating neuroinflammation and microglia polarization. Our previous report reveals activation of GPER1 improves the neurological deficits after ICH via inhibition of A1 astrocytes. However, the role of GPER1 on the protection of WMI and modulation of microglia polarization after ICH remains unclear. METHODS In present study, ICH mice model was induced by autologous whole blood injection and in vitro ICH model was established via treatment BV2 cells with FeSO4. Mice were treated with GPER1 agonist G1, antagonist G15 and BV2 cells were treated with G1, G15 or EGFR inhibitor AG1478. Besides, BV2 conditional medium was used to intervene MO3.13 oligodendrocytes. Immunostaining, immunoblots, transmission electron microscope and PI staining were used to determine the WMI, microglia polarization and potential molecular mechanism after ICH, respectively. RESULTS Our data showed treatment with G1 ameliorated the WMI on the day 3 after ICH. Besides, activation of GPER1 reduced the release of IL-1β, TNF-α and increased the produce of IL-4, IL-10 as well as shifting microglia from proinflammatory M1 to anti-inflammatory M2 phenotype in vivo and in vitro. Meanwhile, MO3.13 cells treated with BV2 conditional medium validated GPER1 alleviated oligodendrocytes death via mitigating neuroinflammation and modulating microglia polarization. Mechanistic study demonstrated EGFR/Stat3 signaling pathway was involved in the protection of WMI and modulation microglia polarization after ICH. CONCLUSION Collectively, our findings demonstrated activation of GPER1 alleviated WMI via modulating microglia M2 polarization after ICH through EGFR/Stat3 signaling pathway.
Collapse
Affiliation(s)
- Xuyang Zhang
- Department of Neurosurgery, Southwest Hospital, The Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| | - Jianchao Mao
- Department of Neurosurgery, Southwest Hospital, The Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| | - Huanhuan Li
- Department of Neurosurgery, Southwest Hospital, The Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| | - Chao Zhang
- Department of Neurosurgery, Southwest Hospital, The Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| | - Hongfei Ge
- Department of Neurosurgery, Southwest Hospital, The Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| | - Jun Zhong
- Department of Neurosurgery, Southwest Hospital, The Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| |
Collapse
|
3
|
Mhaouty-Kodja S, Zalko D, Tait S, Testai E, Viguié C, Corsini E, Grova N, Buratti FM, Cabaton NJ, Coppola L, De la Vieja A, Dusinska M, El Yamani N, Galbiati V, Iglesias-Hernández P, Kohl Y, Maddalon A, Marcon F, Naulé L, Rundén-Pran E, Salani F, Santori N, Torres-Ruiz M, Turner JD, Adamovsky O, Aiello-Holden K, Dirven H, Louro H, Silva MJ. A critical review to identify data gaps and improve risk assessment of bisphenol A alternatives for human health. Crit Rev Toxicol 2024; 54:696-753. [PMID: 39436315 DOI: 10.1080/10408444.2024.2388712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 07/29/2024] [Accepted: 07/30/2024] [Indexed: 10/23/2024]
Abstract
Bisphenol A (BPA), a synthetic chemical widely used in the production of polycarbonate plastic and epoxy resins, has been associated with a variety of adverse effects in humans including metabolic, immunological, reproductive, and neurodevelopmental effects, raising concern about its health impact. In the EU, it has been classified as toxic to reproduction and as an endocrine disruptor and was thus included in the candidate list of substances of very high concern (SVHC). On this basis, its use has been banned or restricted in some products. As a consequence, industries turned to bisphenol alternatives, such as bisphenol S (BPS) and bisphenol F (BPF), which are now found in various consumer products, as well as in human matrices at a global scale. However, due to their toxicity, these two bisphenols are in the process of being regulated. Other BPA alternatives, whose potential toxicity remains largely unknown due to a knowledge gap, have also started to be used in manufacturing processes. The gradual restriction of the use of BPA underscores the importance of understanding the potential risks associated with its alternatives to avoid regrettable substitutions. This review aims to summarize the current knowledge on the potential hazards related to BPA alternatives prioritized by European Regulatory Agencies based on their regulatory relevance and selected to be studied under the European Partnership for the Assessment of Risks from Chemicals (PARC): BPE, BPAP, BPP, BPZ, BPS-MAE, and TCBPA. The focus is on data related to toxicokinetic, endocrine disruption, immunotoxicity, developmental neurotoxicity, and genotoxicity/carcinogenicity, which were considered the most relevant endpoints to assess the hazard related to those substances. The goal here is to identify the data gaps in BPA alternatives toxicology and hence formulate the future directions that will be taken in the frame of the PARC project, which seeks also to enhance chemical risk assessment methodologies using new approach methodologies (NAMs).
Collapse
Affiliation(s)
- Sakina Mhaouty-Kodja
- CNRS UMR 8246, INSERM U1130, Neuroscience Paris Seine - Institut de Biologie Paris Seine, Sorbonne Université, Paris, France
| | - Daniel Zalko
- INRAE, UMR1331 Toxalim (Research Center in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UT3, Toulouse, France
| | - Sabrina Tait
- Center for Gender-Specific Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Emanuela Testai
- Department of Environment and Health, Mechanisms, Biomarkers and Models Unit, Istituto Superiore di Sanità, Rome, Italy
| | - Catherine Viguié
- INRAE, UMR1331 Toxalim (Research Center in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UT3, Toulouse, France
| | - Emanuela Corsini
- Department of Pharmacological and Biomolecular Sciences 'Rodolfo Paoletti', Università degli Studi di Milano - School of Pharmacy, Milan, Italy
| | - Nathalie Grova
- Department of Infection and Immunity, Immune Endocrine Epigenetics Research Group, Luxembourg Institute of Health, Esch-Sur-Alzette, Luxembourg
| | - Franca Maria Buratti
- Department of Environment and Health, Mechanisms, Biomarkers and Models Unit, Istituto Superiore di Sanità, Rome, Italy
| | - Nicolas J Cabaton
- INRAE, UMR1331 Toxalim (Research Center in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UT3, Toulouse, France
| | - Lucia Coppola
- Center for Gender-Specific Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Antonio De la Vieja
- Endocrine Tumor Unit from Chronic Disease Program (UFIEC), Instituto de Salud Carlos III (ISCIII), Majadahonda, Madrid, Spain
| | - Maria Dusinska
- Department for Environmental Chemistry, Health Effects Laboratory, NILU-Norwegian Institute for Air Research, Kjeller, Norway
| | - Naouale El Yamani
- Department for Environmental Chemistry, Health Effects Laboratory, NILU-Norwegian Institute for Air Research, Kjeller, Norway
| | - Valentina Galbiati
- Department of Pharmacological and Biomolecular Sciences 'Rodolfo Paoletti', Università degli Studi di Milano - School of Pharmacy, Milan, Italy
| | - Patricia Iglesias-Hernández
- Endocrine Tumor Unit from Chronic Disease Program (UFIEC), Instituto de Salud Carlos III (ISCIII), Majadahonda, Madrid, Spain
| | - Yvonne Kohl
- Fraunhofer Institute for Biomedical Engineering IBMT, Sulzbach, Germany
| | - Ambra Maddalon
- Department of Pharmacological and Biomolecular Sciences 'Rodolfo Paoletti', Università degli Studi di Milano - School of Pharmacy, Milan, Italy
| | - Francesca Marcon
- Department of Environment and Health, Mechanisms, Biomarkers and Models Unit, Istituto Superiore di Sanità, Rome, Italy
| | - Lydie Naulé
- CNRS UMR 8246, INSERM U1130, Neuroscience Paris Seine - Institut de Biologie Paris Seine, Sorbonne Université, Paris, France
| | - Elise Rundén-Pran
- Department for Environmental Chemistry, Health Effects Laboratory, NILU-Norwegian Institute for Air Research, Kjeller, Norway
| | - Francesca Salani
- Department of Environment and Health, Mechanisms, Biomarkers and Models Unit, Istituto Superiore di Sanità, Rome, Italy
| | - Nicoletta Santori
- Department of Environment and Health, Mechanisms, Biomarkers and Models Unit, Istituto Superiore di Sanità, Rome, Italy
| | - Mónica Torres-Ruiz
- National Center for Environmental Health (CNSA), Instituto de Salud Carlos III (ISCIII), Majadahonda, Madrid, Spain
| | - Jonathan D Turner
- Department of Infection and Immunity, Immune Endocrine Epigenetics Research Group, Luxembourg Institute of Health, Esch-Sur-Alzette, Luxembourg
| | - Ondrej Adamovsky
- Faculty of Science, Masaryk University, RECETOX, Brno, Czech Republic
| | | | - Hubert Dirven
- Department of Chemical Toxicology - Division of Climate and the Environment, Norwegian Institute of Public Health, Oslo, Norway
| | - Henriqueta Louro
- Department of Human Genetics, National Institute of Health Dr. Ricardo Jorge, Lisbon, Portugal
- Centre for Toxicogenomics and Human Health, Nova Medical School, Universidade Nova de Lisboa, Lisbon, Portugal
| | - Maria João Silva
- Department of Human Genetics, National Institute of Health Dr. Ricardo Jorge, Lisbon, Portugal
- Centre for Toxicogenomics and Human Health, Nova Medical School, Universidade Nova de Lisboa, Lisbon, Portugal
| |
Collapse
|
4
|
Batallán Burrowes AA, Moisan É, Garrone A, Buynack LM, Chapman CA. 17β-Estradiol reduces inhibitory synaptic currents in entorhinal cortex neurons through G protein-coupled estrogen receptor-1 activation of extracellular signal-regulated kinase. Hippocampus 2024; 34:454-463. [PMID: 39150316 DOI: 10.1002/hipo.23621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 05/02/2024] [Accepted: 06/05/2024] [Indexed: 08/17/2024]
Abstract
Estrogens are believed to modulate cognitive functions in part through the modulation of synaptic transmission in the cortex and hippocampus. Administration of 17β-estradiol (E2) can rapidly enhance excitatory synaptic transmission in the hippocampus and facilitate excitatory synaptic transmission in rat lateral entorhinal cortex via activation of the G protein-coupled estrogen receptor-1 (GPER1). To assess the mechanisms through which GPER1 activation facilitates synaptic transmission, we assessed the effects of acute 10 nM E2 administration on pharmacologically isolated evoked excitatory and inhibitory synaptic currents in layer II/III entorhinal neurons. Female Long-Evans rats were ovariectomized between postnatal day (PD) 63 and 74 and implanted with a subdermal E2 capsule to maintain continuous low levels of E2. Electrophysiological recordings were obtained between 7 and 20 days after ovariectomy. Application of E2 for 20 min did not significantly affect AMPA or NMDA receptor-mediated excitatory synaptic currents. However, GABA receptor-mediated inhibitory synaptic currents (IPSCs) were markedly reduced by E2 and returned towards baseline levels during the 20-min washout period. The inhibition of GABA-mediated IPSCs was blocked in the presence of the GPER1 receptor antagonist G15. GPER1 can modulate protein kinase A (PKA), but blocking PKA with intracellular KT5720 did not prevent the E2-induced reduction in IPSCs. GPER1 can also stimulate extracellular signal-regulated kinase (ERK), a negative modulator of GABAA receptors, and blocking activation of ERK with PD90859 prevented the E2-induced reduction of IPSCs. E2 can therefore result in a rapid GPER1 and ERK signaling-mediated reduction in GABA-mediated IPSCs. This provides a novel mechanism through which E2 can rapidly modulate synaptic excitability in entorhinal layer II/III neurons and may also contribute to E2 and ERK-dependent alterations in synaptic transmission in other brain areas.
Collapse
Affiliation(s)
- Ariel A Batallán Burrowes
- Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, Montreal, Québec, Canada
| | - Élyse Moisan
- Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, Montreal, Québec, Canada
| | - Aurelie Garrone
- Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, Montreal, Québec, Canada
| | - Lauren M Buynack
- Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, Montreal, Québec, Canada
| | - C Andrew Chapman
- Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, Montreal, Québec, Canada
| |
Collapse
|
5
|
Proffitt MR, Smith GT. Species variation in steroid hormone-related gene expression contributes to species diversity in sexually dimorphic communication in electric fishes. Horm Behav 2024; 164:105576. [PMID: 38852479 PMCID: PMC11330740 DOI: 10.1016/j.yhbeh.2024.105576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 05/08/2024] [Accepted: 05/31/2024] [Indexed: 06/11/2024]
Abstract
Sexually dimorphic behaviors are often regulated by gonadal steroid hormones. Species diversity in behavioral sex differences may arise as expression of genes mediating steroid action in brain regions controlling these behaviors evolves. The electric communication signals of apteronotid knifefishes are an excellent model for comparatively studying neuroendocrine regulation of sexually dimorphic behavior. These fish produce and detect weak electric organ discharges (EODs) for electrolocation and communication. EOD frequency (EODf), controlled by the medullary pacemaker nucleus (Pn), is sexually dimorphic and regulated by androgens and estrogens in some species, but is sexually monomorphic and unaffected by hormones in other species. We quantified expression of genes for steroid receptors, metabolizing enzymes, and cofactors in the Pn of two species with sexually dimorphic EODf (Apteronotus albifrons and Apteronotus leptorhynchus) and two species with sexually monomorphic EODf ("Apteronotus" bonapartii and Parapteronotus hasemani). The "A." bonapartii Pn expressed lower levels of androgen receptor (AR) genes than the Pn of species with sexually dimorphic EODf. In contrast, the P. hasemani Pn robustly expressed AR genes, but expressed lower levels of genes for 5α-reductases, which convert androgens to more potent metabolites, and higher levels of genes for 17β-hydroxysteroid dehydrogenases that oxidize androgens and estrogens to less potent forms. These findings suggest that sexual monomorphism of EODf arose convergently via two different mechanisms. In "A." bonapartii, reduced Pn expression of ARs likely results in insensitivity of EODf to androgens, whereas in P. hasemani, gonadal steroids may be metabolically inactivated in the Pn, reducing their potential to influence EODf.
Collapse
Affiliation(s)
- Melissa R Proffitt
- Department of Biology, Indiana University, 1001 E. 3(rd) St., Bloomington, IN 47405, USA; Center for the Integrative Study of Animal Behavior, Indiana University, 409 N. Park Ave., Bloomington, IN 47505, USA
| | - G Troy Smith
- Department of Biology, Indiana University, 1001 E. 3(rd) St., Bloomington, IN 47405, USA; Center for the Integrative Study of Animal Behavior, Indiana University, 409 N. Park Ave., Bloomington, IN 47505, USA.
| |
Collapse
|
6
|
Grubić Kezele T, Omrčen H, Batičić L, Šućurović S, Zoričić Cvek S. Joint Inflammation Correlates with Joint GPR30 Expression in Males and Hippocampal GPR30 Expression in Females in a Rat Model of Rheumatoid Arthritis. Int J Mol Sci 2024; 25:7864. [PMID: 39063107 PMCID: PMC11277240 DOI: 10.3390/ijms25147864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 07/12/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024] Open
Abstract
It is not entirely clear how the interaction between joint inflammation and the central nervous system (CNS) response in rheumatoid arthritis (RA) works, and what pathophysiology underlies the sex differences in coexisting neuropsychiatric comorbidities. It is known that estrogen hormones reduce inflammation in RA and that this occurs mainly via the stimulation of G protein-coupled receptor-30 (GPR30), also known as G protein-coupled estrogen receptor (GPER) 1. However, changes in GPR30 expression and sex differences induced by local and systemic inflammation in RA are not yet known. Our aim was to reveal sex differences in the expression and association of joint GPR30 with local and systemic inflammation, clinical course and furthermore with hippocampal GPR30 expression during pristane-induced arthritis (PIA) in Dark Agouti (DA) rats, an animal model of RA. Furthermore, we demonstrated sex-specific differences in the association between joint and systemic inflammation and hippocampal microglia during PIA. Our results suggest sex-specific differences not only in the clinical course and serum levels of pro-inflammatory cytokines but also in the expression of GPR30. Female rats show greater synovial inflammation and greater damage to the articular cartilage compared to males during PIA attack. Male rats express higher levels of synovial and cartilaginous GPR30 than females during PIA, which correlates with a less severe clinical course. The correlation between synovial and cartilaginous GPR30 and joint inflammation scores (Krenn and Mankin) in male rats suggests that the more severe the joint inflammation, the higher the GPR30 expression. At the same time, there is no particular upregulation of hippocampal GPR30 in males. On the other hand, female rats express higher levels of neuroprotective GPR30 in the hippocampus than male rats at the basic level and during PIA attack. In addition, females have a higher number of Iba-1+ cells in the hippocampus during PIA attack that strongly correlates with the clinical score, serum levels of IL-17A, and Krenn and Mankin scores. These results suggest that male rats are better protected from inflammation in the joints and female rats are better protected from the inflammation in the hippocampus during a PIA attack, independently of microglia proliferation. However, in the remission phase, synovial GPR30 expression suddenly increases in female rats, as does hippocampal GPR30 expression in males. Further experiments with a longer remission period are needed to investigate the molecular background of these sex differences, as well as microglia phenotype profiling.
Collapse
MESH Headings
- Animals
- Female
- Male
- Receptors, G-Protein-Coupled/metabolism
- Receptors, G-Protein-Coupled/genetics
- Arthritis, Rheumatoid/metabolism
- Arthritis, Rheumatoid/pathology
- Arthritis, Rheumatoid/genetics
- Hippocampus/metabolism
- Rats
- Disease Models, Animal
- Inflammation/metabolism
- Arthritis, Experimental/metabolism
- Arthritis, Experimental/pathology
- Sex Characteristics
- Receptors, Estrogen/metabolism
- Receptors, Estrogen/genetics
- Microglia/metabolism
- Sex Factors
- Terpenes
Collapse
Affiliation(s)
- Tanja Grubić Kezele
- Department of Physiology, Immunology and Pathophysiology, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
- Clinical Department for Clinical Microbiology, Clinical Hospital Centre Rijeka, 51000 Rijeka, Croatia
| | - Hrvoje Omrčen
- Department of Microbiology, Teaching Institute of Public Health of Primorje-Gorski Kotar County, 51000 Rijeka, Croatia;
| | - Lara Batičić
- Department of Medical Chemistry, Biochemistry and Clinical Chemistry, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia;
| | - Sandra Šućurović
- Specialized Hematology Laboratory, Medical Centre Ljubljana, 1000 Ljubljana, Slovenia;
| | - Sanja Zoričić Cvek
- Department of Anatomy, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia;
| |
Collapse
|
7
|
Zhang Y, Tan X, Tang C. Estrogen-immuno-neuromodulation disorders in menopausal depression. J Neuroinflammation 2024; 21:159. [PMID: 38898454 PMCID: PMC11188190 DOI: 10.1186/s12974-024-03152-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Accepted: 06/11/2024] [Indexed: 06/21/2024] Open
Abstract
A significant decrease in estrogen levels puts menopausal women at high risk for major depression, which remains difficult to cure despite its relatively clear etiology. With the discovery of abnormally elevated inflammation in menopausal depressed women, immune imbalance has become a novel focus in the study of menopausal depression. In this paper, we examined the characteristics and possible mechanisms of immune imbalance caused by decreased estrogen levels during menopause and found that estrogen deficiency disrupted immune homeostasis, especially the levels of inflammatory cytokines through the ERα/ERβ/GPER-associated NLRP3/NF-κB signaling pathways. We also analyzed the destruction of the blood-brain barrier, dysfunction of neurotransmitters, blockade of BDNF synthesis, and attenuation of neuroplasticity caused by inflammatory cytokine activity, and investigated estrogen-immuno-neuromodulation disorders in menopausal depression. Current research suggests that drugs targeting inflammatory cytokines and NLRP3/NF-κB signaling molecules are promising for restoring homeostasis of the estrogen-immuno-neuromodulation system and may play a positive role in the intervention and treatment of menopausal depression.
Collapse
Affiliation(s)
- Yuling Zhang
- College of Life Science, Henan Normal University, Xinxiang, 453007, Henan, China
| | - Xiying Tan
- Department of Neurology, Xinxiang City First People's Hospital, Xinxiang, 453000, Henan, China
| | - Chaozhi Tang
- College of Life Science, Henan Normal University, Xinxiang, 453007, Henan, China.
| |
Collapse
|
8
|
Mao J, Guo Y, Li H, Ge H, Zhang C, Feng H, Zhong J, Hu R, Wang X. Modulation of GPER1 alleviates early brain injury via inhibition of A1 reactive astrocytes activation after intracerebral hemorrhage in mice. Heliyon 2024; 10:e26909. [PMID: 38439827 PMCID: PMC10909704 DOI: 10.1016/j.heliyon.2024.e26909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 02/20/2024] [Accepted: 02/21/2024] [Indexed: 03/06/2024] Open
Abstract
Background Early brain injury (EBI) caused by inflammatory responses in acute phase of Intracerebral hemorrhage (ICH) plays a vital role in the pathological progression of ICH. Increasing evidences demonstrate A1 reactive astrocytes are associated with the severity of EBI. G-protein coupled estrogen receptor 1 (GPER1) has been proved mediating the neuroprotective effects of estrogen in central nervous system (CNS) disease. However, whether GPER1 plays a protective effect on ICH and A1 reactive astrocytes activation is not well studied. Methods ICH model was established by infused the autologous whole blood into the right basal ganglia in wild type and GPER1 knockout mice. GPER1 specific agonist G1 and antagonist G15 were administered by intraperitoneal injection at 1 h or 0.5 h after ICH. Neurological function was detected on day 1 and day 3 by open field test and corner turn test following ICH. Besides, A1 reactive astrocytes were determined by immunofluorescence staining after ICH on day 3. To further identify the possible mechanism of GPER1 mediated neuroprotective effect, Western blot assays was performed after ICH on day 3. Results After ICH, G1 treatment alleviated mice neurobehavior deficits on day 1 and day 3. Meanwhile, G1 treatment also significantly reduced the GFAP positive astrocytes and the C3 positive cells after ICH. Interestingly, G15 reversed the protective effect of G1 on the neurobehavior of ICH mice. Meanwhile, the expression of GFAP+C3+ A1 reactive astrocytes were also reduced by activation of GPER1. Mechanistic studies indicated TLR4 and NF-κB mediated the neuroprotective effect of GPER1. Conclusion Generally, activation of GPER1 alleviated the EBI through inhibiting A1 reactive astrocytes activation via TLR4/NF-κB pathway after ICH in mice. Additionally, GPER1may be a promising target for ICH treatment.
Collapse
Affiliation(s)
- Jianchao Mao
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450052, China
- Department of Neurosurgery, Southwest Hospital, The Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Yongkun Guo
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450052, China
| | - Huanhuan Li
- Department of Neurosurgery, Southwest Hospital, The Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Hongfei Ge
- Department of Neurosurgery, Southwest Hospital, The Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Chao Zhang
- Department of Neurosurgery, Southwest Hospital, The Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Hua Feng
- Department of Neurosurgery, Southwest Hospital, The Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Jun Zhong
- Department of Neurosurgery, Southwest Hospital, The Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Rong Hu
- Department of Neurosurgery, Southwest Hospital, The Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Xinjun Wang
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450052, China
| |
Collapse
|
9
|
Hodes GE, Bangasser D, Sotiropoulos I, Kokras N, Dalla C. Sex Differences in Stress Response: Classical Mechanisms and Beyond. Curr Neuropharmacol 2024; 22:475-494. [PMID: 37855285 PMCID: PMC10845083 DOI: 10.2174/1570159x22666231005090134] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 07/28/2023] [Accepted: 08/09/2023] [Indexed: 10/20/2023] Open
Abstract
Neuropsychiatric disorders, which are associated with stress hormone dysregulation, occur at different rates in men and women. Moreover, nowadays, preclinical and clinical evidence demonstrates that sex and gender can lead to differences in stress responses that predispose males and females to different expressions of similar pathologies. In this curated review, we focus on what is known about sex differences in classic mechanisms of stress response, such as glucocorticoid hormones and corticotrophin-releasing factor (CRF), which are components of the hypothalamicpituitary- adrenal (HPA) axis. Then, we present sex differences in neurotransmitter levels, such as serotonin, dopamine, glutamate and GABA, as well as indices of neurodegeneration, such as amyloid β and Tau. Gonadal hormone effects, such as estrogens and testosterone, are also discussed throughout the review. We also review in detail preclinical data investigating sex differences caused by recentlyrecognized regulators of stress and disease, such as the immune system, genetic and epigenetic mechanisms, as well neurosteroids. Finally, we discuss how understanding sex differences in stress responses, as well as in pharmacology, can be leveraged into novel, more efficacious therapeutics for all. Based on the supporting evidence, it is obvious that incorporating sex as a biological variable into preclinical research is imperative for the understanding and treatment of stress-related neuropsychiatric disorders, such as depression, anxiety and Alzheimer's disease.
Collapse
Affiliation(s)
| | - Debra Bangasser
- Center for Behavioral Neuroscience, Georgia State University, Atlanta, GA, USA
| | - Ioannis Sotiropoulos
- Institute of Biosciences & Applications NCSR “Demokritos”, Athens, Greece
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
| | - Nikolaos Kokras
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
- First Department of Psychiatry, Eginition Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Christina Dalla
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
10
|
Upadhayay S, Gupta R, Singh S, Mundkar M, Singh G, Kumar P. Involvement of the G-Protein-Coupled Estrogen Receptor-1 (GPER) Signaling Pathway in Neurodegenerative Disorders: A Review. Cell Mol Neurobiol 2023; 43:1833-1847. [PMID: 36307605 PMCID: PMC11412173 DOI: 10.1007/s10571-022-01301-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 10/18/2022] [Indexed: 11/26/2022]
Abstract
The G-protein-coupled estrogen receptor-1 (GPER) is an extranuclear estrogen receptor that regulates the expression of several downstream signaling pathways with a variety of biological actions including cell migration, proliferation, and apoptosis in different parts of the brain area. It is endogenously activated by estrogen, a steroidal hormone that binds to GPER receptors which help in maintaining cellular homeostasis and neuronal integrity as well as influences neurogenesis. In contrast, neurodegenerative disorders are a big problem for society, and still many people suffer from motor and cognitive impairments. Research to date reported that GPER has the potential to whittle down motor abnormalities and cognitive dysfunction by limiting the progression of neurodegenerative disorders. Although several findings suggest that GPER activation accelerated transcription of the PI3K/Akt/Gsk-3β and ERK1/2 signaling pathway that halt disease progression by decreasing oxidative stress, neuroinflammation, and apoptosis. Accordingly, the goal of this review is to highlight the basic mechanism of GPER signaling pathway-mediated neuroprotection in various neurodegenerative disorders including Parkinson's disease (PD), Huntington's disease (HD), Tardive dyskinesia (TD), and Epilepsy. This review also discusses the role of the GPER activators which might be a promising therapeutic target option to treat neurodegenerative disorders. All the data were obtained from published articles in PubMed (353), Web of Science (788), and Scopus (770) databases using the search terms: GPER, PD, HD, TD, epilepsy, and neurodegenerative disorders.
Collapse
Affiliation(s)
- Shubham Upadhayay
- Department of Pharmacology, Central University of Punjab, Ghudda Campus, Ghudda, Bathinda, Punjab, 151401, India
| | - Rishav Gupta
- Department of Pharmacology, Central University of Punjab, Ghudda Campus, Ghudda, Bathinda, Punjab, 151401, India
| | - Surbhi Singh
- Department of Pharmacology, Central University of Punjab, Ghudda Campus, Ghudda, Bathinda, Punjab, 151401, India
| | - Maroti Mundkar
- Department of Pharmacology, Central University of Punjab, Ghudda Campus, Ghudda, Bathinda, Punjab, 151401, India
| | - Gursewak Singh
- Department of Pharmacology, Central University of Punjab, Ghudda Campus, Ghudda, Bathinda, Punjab, 151401, India
| | - Puneet Kumar
- Department of Pharmacology, Central University of Punjab, Ghudda Campus, Ghudda, Bathinda, Punjab, 151401, India.
| |
Collapse
|
11
|
Hameed RA, Ahmed EK, Mahmoud AA, Atef AA. G protein-coupled estrogen receptor (GPER) selective agonist G1 attenuates the neurobehavioral, molecular and biochemical alterations induced in a valproic acid rat model of autism. Life Sci 2023:121860. [PMID: 37331505 DOI: 10.1016/j.lfs.2023.121860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 03/31/2023] [Accepted: 06/11/2023] [Indexed: 06/20/2023]
Abstract
AIMS Autism Spectrum Disorder (ASD) is a neurodevelopmental disorder with a rising prevalence in boys rather than girls. G protein-coupled estrogen receptor (GPER) activation by its agonist G1 showed a neuroprotective effect, similar to estradiol. The present study aimed to examine the potential of the selective GPER agonist G1 therapy on the behavioral, histopathological, biochemical, and molecular alterations induced in a valproic acid (VPA)-rat model of autism. MAIN METHODS VPA (500 mg/kg) was intraperitoneally administered to female Wistar rats (on gestational day 12.5) to induce the VPA-rat model of autism. The male offspring were intraperitoneally administered with G1 (10 and 20 μg/kg) for 21 days. After the treatment process, rats performed behavioral assessments. Then, sera and hippocampi were collected for biochemical and histopathological examinations and gene expression analysis. KEY FINDINGS GPER agonist G1 attenuated behavioral deficits, including hyperactivity, declined spatial memory and social preferences, anxiety, and repetitive behavior in VPA rats. G1 improved neurotransmission and reduced oxidative stress and histological alteration in the hippocampus. G1 reduced serum free T levels and interleukin-1β and up-regulated GPER, RORα, and aromatase gene expression levels in the hippocampus. SIGNIFICANCE The present study suggests that activation of GPER by its selective agonist G1 altered the derangements induced in a VPA-rat model of autism. G1 normalized free T levels via up-regulation of hippocampal RORα and aromatase gene expression. G1 provoked estradiol neuroprotective functions via up-regulation of hippocampal GPER expression. The G1 treatment and GPER activation provide a promising therapeutic approach to counteract the autistic-like symptoms.
Collapse
Affiliation(s)
- Rehab Abdel Hameed
- Department of Biochemistry, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Emad K Ahmed
- Department of Biochemistry, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Asmaa A Mahmoud
- Department of Zoology, Faculty of Science, Ain Shams University, Cairo, Egypt.
| | - Azza A Atef
- Department of Biochemistry, Faculty of Science, Ain Shams University, Cairo, Egypt
| |
Collapse
|
12
|
G-protein coupled estrogen receptor (GPER1) activation promotes synaptic insertion of AMPA receptors and induction of chemical LTP at hippocampal temporoammonic-CA1 synapses. Mol Brain 2023; 16:16. [PMID: 36709268 PMCID: PMC9883958 DOI: 10.1186/s13041-023-01003-3] [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: 09/12/2022] [Accepted: 01/12/2023] [Indexed: 01/30/2023] Open
Abstract
It is well documented that 17β estradiol (E2) regulates excitatory synaptic transmission at hippocampal Shaffer-collateral (SC)-CA1 synapses, via activation of the classical estrogen receptors (ERα and ERβ). Hippocampal CA1 pyramidal neurons are also innervated by the temporoammonic (TA) pathway, and excitatory TA-CA1 synapses are reported to be regulated by E2. Recent studies suggest a role for the novel G-protein coupled estrogen receptor (GPER1) at SC-CA1 synapses, however, the role of GPER1 in mediating the effects of E2 at juvenile TA-CA1 synapses is unclear. Here we demonstrate that the GPER1 agonist, G1 induces a persistent, concentration-dependent (1-10 nM) increase in excitatory synaptic transmission at TA-CA1 synapses and this effect is blocked by selective GPER1 antagonists. The ability of GPER1 to induce this novel form of chemical long-term potentiation (cLTP) was prevented following blockade of N-methyl-D-aspartate (NMDA) receptors, and it was not accompanied by any change in paired pulse facilitation ratio (PPR). GPER1-induced cLTP involved activation of ERK but was independent of phosphoinositide 3-kinase (PI3K) signalling. Prior treatment with philanthotoxin prevented the effects of G1, indicating that synaptic insertion of GluA2-lacking α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors underlies GPER1-induced cLTP. Furthermore, activity-dependent LTP occluded G1-induced cLTP and vice versa, indicating that these processes have overlapping expression mechanisms. Activity-dependent LTP was blocked by the GPER1 antagonist, G15, suggesting that GPER1 plays a role in NMDA-dependent LTP at juvenile TA-CA1 synapses. These findings add a new dimension to our understanding of GPER1 in modulating neuronal plasticity with relevance to age-related neurodegenerative conditions.
Collapse
|
13
|
Abstract
Depression and anxiety disorders carry a tremendous worldwide burden and emerge as a significant cause of disability among western societies. Both disorders are known to disproportionally affect women, as they are twice more likely to be diagnosed and moreover, they are also prone to suffer from female-specific mood disorders. Importantly, the prevalence of these affective disorders has notably risen after the COVID pandemic, especially in women. In this chapter, we describe factors that are possibly contributing to the expression of such sex differences in depression and anxiety. For this, we overview the effect of transcriptomic and genetic factors, the immune system, neuroendocrine aspects, and cognition. Furthermore, we also provide evidence of sex differences in antidepressant response and their causes. Finally, we emphasize the importance to consider sex as a biological variable in preclinical and clinical research, which may facilitate the discovery and development of new and more efficacious antidepressant and anxiolytic pharmacotherapies for both women and men.
Collapse
Affiliation(s)
- Pavlina Pavlidi
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Nikolaos Kokras
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
- First Department of Psychiatry, Eginition Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Christina Dalla
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece.
| |
Collapse
|
14
|
Luo W, Yan Y, Cao Y, Zhang Y, Zhang Z. The effects of GPER on age-associated memory impairment induced by decreased estrogen levels. Front Mol Biosci 2023; 10:1097018. [PMID: 37021109 PMCID: PMC10069632 DOI: 10.3389/fmolb.2023.1097018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Accepted: 02/08/2023] [Indexed: 04/07/2023] Open
Abstract
Estrogen, as a pleiotropic endocrine hormone, not only regulates the physiological functions of peripheral tissues but also exerts vital neuroregulatory effects in the central nervous system (CNS), such as the development of neurons and the formation of neural network connections, wherein rapid estrogen-mediated reactions positively stimulate spinogenesis and regulate synaptic plasticity and synaptic transmission to facilitate cognitive and memory performance. These fast non-genomic effects can be initiated by membrane-bound estrogen receptors (ERs), three best known of which are ERα, ERβ, and G protein-coupled estrogen receptor (GPER). To date, the effects of ERα and ERβ have been well studied in age-associated memory impairment, whereas there is still a lack of attention to the role of GPER in age-associated memory impairment, and there are still disputes about whether GPER indeed functions as an ER to enhance learning and memory. In this review, we provide a systematic overview of the role of GPER in age-associated memory impairment based on its expression, distribution, and signaling pathways, which might bring some inspiration for translational drugs targeting GPER for age-related diseases and update knowledge on the role of estrogen and its receptor system in the brain.
Collapse
Affiliation(s)
- Wenyu Luo
- Department of Ultrasound, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yudie Yan
- Department of Ultrasound, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yunpeng Cao
- Department of Neurology, The First Hospital of China Medical University, Shenyang, Liaoning, China
- *Correspondence: Zhen Zhang, ; Yunpeng Cao, ; Yanbo Zhang,
| | - Yanbo Zhang
- Department of Psychiatry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
- *Correspondence: Zhen Zhang, ; Yunpeng Cao, ; Yanbo Zhang,
| | - Zhen Zhang
- Department of Ultrasound, The First Hospital of China Medical University, Shenyang, Liaoning, China
- *Correspondence: Zhen Zhang, ; Yunpeng Cao, ; Yanbo Zhang,
| |
Collapse
|
15
|
Poirier AA, Côté M, Bourque M, Jarras H, Lamontagne-Proulx J, Morissette M, Paolo TD, Soulet D. DIFFERENTIAL CONTRIBUTION OF ESTROGEN RECEPTORS TO THE INTESTINAL THERAPEUTIC EFFECTS OF 17β-ESTRADIOL IN A MURINE MODEL OF PARKINSON'S DISEASE. Brain Res Bull 2022; 187:85-97. [PMID: 35781029 DOI: 10.1016/j.brainresbull.2022.06.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 06/07/2022] [Accepted: 06/29/2022] [Indexed: 11/19/2022]
Abstract
Beneficial effects of estrogens have been reported in Parkinson's disease (PD) for many years. We previously reported their neuroprotective and anti-inflammatory potentials in the enteric nervous system of the intestine, a region possibly affected during the early stages of the disease according to Braak's hypothesis. Three different estrogen receptors have been characterized to date: the estrogen receptor alpha (ERα), the estrogen receptor beta (ERβ) and the G protein coupled estrogen receptor 1 (GPER1). The aim of the present study was to decipher the individual contribution of each estrogen receptor to the therapeutic properties of 17β-estradiol (E2) in the myenteric plexus of the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of PD. Different agonists, 4,4',4''-(4-propyl-[1H]-pyrazole-1,3,5-triyl)trisphenol (PPT; ERα), 2,3-bis(4-hydroxyphenyl)-propionitrile (DPN; ERβ), G1 (GPER1), and antagonists, ICI 182,780 (ERα and ERβ), G15 (GPER1), were used to analyze the involvement of each receptor. We confirmed that G1 protects dopamine (DA) neurons to a similar extent as E2. An anti-inflammatory effect on proinflammatory macrophages and cultured human monocytes was also demonstrated with E2 and G1. The effects of PPT and DPN were less potent than G1 with only a partial neuroprotection of DA neurons by PPT and a partial reduction of interleukin (IL)-1β production in monocytes by PPT and DPN. Overall, the present results indicate that the positive outcomes of estrogens are mainly through activation of GPER1. Therefore, this suggests that targeting GPER1 could be a promising approach for future estrogen-based hormone therapies during early PD.
Collapse
Affiliation(s)
- Andrée-Anne Poirier
- Centre de recherche du CHU de Québec, Québec, QC, Canada; Faculté de pharmacie, Université Laval, Québec, QC, Canada
| | - Mélissa Côté
- Centre de recherche du CHU de Québec, Québec, QC, Canada
| | | | - Hend Jarras
- Centre de recherche du CHU de Québec, Québec, QC, Canada; Faculté de pharmacie, Université Laval, Québec, QC, Canada
| | - Jérôme Lamontagne-Proulx
- Centre de recherche du CHU de Québec, Québec, QC, Canada; Faculté de pharmacie, Université Laval, Québec, QC, Canada
| | | | - Thérèse Di Paolo
- Centre de recherche du CHU de Québec, Québec, QC, Canada; Faculté de pharmacie, Université Laval, Québec, QC, Canada
| | - Denis Soulet
- Centre de recherche du CHU de Québec, Québec, QC, Canada; Faculté de pharmacie, Université Laval, Québec, QC, Canada; Institut sur la Nutrition et les Aliments Fonctionnels (INAF), Université Laval, Québec, QC, Canada.
| |
Collapse
|
16
|
Trouillet AC, Ducroq S, Naulé L, Capela D, Parmentier C, Radovick S, Hardin-Pouzet H, Mhaouty-Kodja S. Deletion of neural estrogen receptor alpha induces sex differential effects on reproductive behavior in mice. Commun Biol 2022; 5:383. [PMID: 35444217 PMCID: PMC9021208 DOI: 10.1038/s42003-022-03324-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Accepted: 03/25/2022] [Indexed: 11/20/2022] Open
Abstract
Estrogen receptor (ER) α is involved in several estrogen-modulated neural and peripheral functions. To determine its role in the expression of female and male reproductive behavior, a mouse line lacking the ERα in the nervous system was generated. Mutant females did not exhibit sexual behavior despite normal olfactory preference, and had a reduced number of progesterone receptor-immunoreactive neurons in the ventromedial hypothalamus. Mutant males displayed a moderately impaired sexual behavior and unaffected fertility, despite evidences of altered organization of sexually dimorphic populations in the preoptic area. In comparison, males deleted for both neural ERα and androgen receptor (AR) displayed greater sexual deficiencies. Thus, these data highlight a predominant role for neural ERα in females and a complementary role with the AR in males in the regulation of sexual behavior, and provide a solid background for future analyses of neuronal versus glial implication of these signaling pathways in both sexes. Neural deletion of the estrogen receptor, ERα, inhibits sexual behavior in female mice, but only has moderately effect in male mice. These results contrast with previous studies using global ERα knockouts, which found that ERα is mandatory for reproductive behavior in both sexes.
Collapse
Affiliation(s)
- Anne-Charlotte Trouillet
- Sorbonne Université, CNRS, INSERM, Neuroscience Paris Seine - Institut de Biologie Paris Seine, 75005, Paris, France
| | - Suzanne Ducroq
- Sorbonne Université, CNRS, INSERM, Neuroscience Paris Seine - Institut de Biologie Paris Seine, 75005, Paris, France
| | - Lydie Naulé
- Sorbonne Université, CNRS, INSERM, Neuroscience Paris Seine - Institut de Biologie Paris Seine, 75005, Paris, France
| | - Daphné Capela
- Sorbonne Université, CNRS, INSERM, Neuroscience Paris Seine - Institut de Biologie Paris Seine, 75005, Paris, France
| | - Caroline Parmentier
- Sorbonne Université, CNRS, INSERM, Neuroscience Paris Seine - Institut de Biologie Paris Seine, 75005, Paris, France
| | - Sally Radovick
- Unit of Pediatric Endocrinology, Department of Pediatrics, Rutgers-Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Hélène Hardin-Pouzet
- Sorbonne Université, CNRS, INSERM, Neuroscience Paris Seine - Institut de Biologie Paris Seine, 75005, Paris, France
| | - Sakina Mhaouty-Kodja
- Sorbonne Université, CNRS, INSERM, Neuroscience Paris Seine - Institut de Biologie Paris Seine, 75005, Paris, France.
| |
Collapse
|
17
|
DeLeon C, Pemberton K, Green M, Kalajdzic V, Rosato M, Xu F, Arnatt C. Novel GPER Agonist, CITFA, Increases Neurite Growth in Rat Embryonic (E18) Hippocampal Neurons. ACS Chem Neurosci 2022; 13:1119-1128. [PMID: 35353510 DOI: 10.1021/acschemneuro.1c00811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Numerous studies have reported neuroprotective and procognitive effects of estrogens. The estrogen 17β-estradiol (E2) activates both the classical nuclear estrogen receptors ERα and ERβ as well as the G protein-coupled estrogen receptor (GPER). The differential effects of targeting the classical estrogen receptors over GPER are not well-understood. A limited number of selective GPER compounds have been described. In this study, 10 novel compounds were synthesized and exhibited half-maximal effective concentration values greater than the known GPER agonist G-1 in calcium mobilization assays performed in nonadherent HL-60 cells. Of these compounds, 2-cyclohexyl-4-isopropyl-N-((5-(tetrahydro-2H-pyran-2-yl)furan-2-yl)methyl)aniline, referred to as CITFA, significantly increased axonal and dendritic growth in neurons extracted from embryonic day 18 (E18) fetal rat hippocampal neurons. Confirmation of the results was performed by treating E18 hippocampal neurons with known GPER-selective antagonist G-36 and challenging with either E2, G-1, or CITFA. Results from these studies revealed an indistinguishable difference in neurite outgrowth between the treatment and control groups, exhibiting that neurite outgrowth in response to G-1 and CITFA originates from GPER activation and can be abolished with pretreatment of an antagonist. Subsequent docking studies using a homology model of GPER showed unique docking poses between G-1 and CIFTA. While docking poses differed between the ligands, CIFTA exhibited more favorable distance, bond angle, and strain for hydrogen-bonding and hydrophobic interactions.
Collapse
Affiliation(s)
- Chelsea DeLeon
- The Department of Chemistry, Saint Louis University, St. Louis, Missouri 63103, United States
| | - Kyle Pemberton
- The Department of Biology, College of Arts and Sciences, Saint Louis University, St. Louis, Missouri 63103, United States
- The Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University, St. Louis, Missouri 63104, United States
| | - Michael Green
- The Department of Chemistry, Saint Louis University, St. Louis, Missouri 63103, United States
| | - Vanja Kalajdzic
- The Department of Chemistry, Saint Louis University, St. Louis, Missouri 63103, United States
| | - Martina Rosato
- The Department of Biology, College of Arts and Sciences, Saint Louis University, St. Louis, Missouri 63103, United States
- The Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University, St. Louis, Missouri 63104, United States
| | - Fenglian Xu
- The Department of Biology, College of Arts and Sciences, Saint Louis University, St. Louis, Missouri 63103, United States
- The Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University, St. Louis, Missouri 63104, United States
- The Department of Pharmacology and Physiology, Saint Louis University, St. Louis, Missouri 63103, United States
| | - Christopher Arnatt
- The Department of Chemistry, Saint Louis University, St. Louis, Missouri 63103, United States
- The Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University, St. Louis, Missouri 63104, United States
- The Department of Pharmacology and Physiology, Saint Louis University, St. Louis, Missouri 63103, United States
| |
Collapse
|
18
|
Dennison JL, Volmar CH, Ke D, Wang J, Gravel E, Hammond-Vignini S, Li Z, Timmons JA, Lohse I, Hayward MA, Brothers SP, Wahlestedt C. JOTROL, a Novel Formulation of Resveratrol, Shows Beneficial Effects in the 3xTg-AD Mouse Model. J Alzheimers Dis 2022; 86:173-190. [PMID: 35034905 DOI: 10.3233/jad-215370] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) has minimally effective treatments currently. High concentrations of resveratrol, a polyphenol antioxidant found in plants, have been reported to affect several AD-related and neuroprotective genes. To address the low bioavailability of resveratrol, we investigated a novel oral formulation of resveratrol, JOTROL™, that has shown increased pharmacokinetic properties compared to non-formulated resveratrol in animals and in humans. OBJECTIVE We hypothesized that equivalent doses of JOTROL, compared to non-formulated resveratrol, would result in greater brain exposure to resveratrol, and more efficacious responses on AD biomarkers. METHODS For sub-chronic reversal studies, 15-month-old male triple transgenic (APPSW/PS1M146V/TauP301L; 3xTg-AD) AD mice were treated orally with vehicle or 50 mg/kg JOTROL for 36 days. For prophylactic studies, male and female 3xTg-AD mice were similarly administered vehicle, 50 mg/kg JOTROL, or 50 mg/kg resveratrol for 9 months starting at 4 months of age. A behavioral battery was run, and mRNA and protein from brain and blood were analyzed for changes in AD-related gene and protein expression. RESULTS JOTROL displays significantly increased bioavailability over non-formulated resveratrol. Treatment with JOTROL resulted in AD-related gene expression changes (Adam10, Bace1, Bdnf, Psen1) some of which were brain region-dependent and sex-specific, as well as changes in inflammatory gene and cytokine levels. CONCLUSION JOTROL may be effective as a prophylaxis and/or treatment for AD through increased expression and/or activation of neuroprotective genes, suppression of pro-inflammatory genes, and regulation of central and peripheral cytokine levels.
Collapse
Affiliation(s)
- Jessica L Dennison
- Department of Psychiatry & Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL, USA.,Center for Therapeutic Innovation, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Claude-Henry Volmar
- Department of Psychiatry & Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL, USA.,Center for Therapeutic Innovation, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Danbing Ke
- KDM Laboratories Inc., Montreal, QC, Canada
| | - James Wang
- KDM Laboratories Inc., Montreal, QC, Canada
| | | | | | - Zuomei Li
- NuChem Sciences Inc., St. Laurent, QC, Canada
| | | | - Ines Lohse
- Department of Psychiatry & Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL, USA.,Center for Therapeutic Innovation, University of Miami Miller School of Medicine, Miami, FL, USA
| | | | - Shaun P Brothers
- Department of Psychiatry & Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL, USA.,Center for Therapeutic Innovation, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Claes Wahlestedt
- Department of Psychiatry & Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL, USA.,Center for Therapeutic Innovation, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
19
|
Khaksari M, Hajializadeh Z, Mahani SE, Soltani Z, Asadikaram G. Estrogen receptor agonists induce anti‑edema effects by altering α and β estrogen receptor gene expression. Acta Neurobiol Exp (Wars) 2021; 81:286-294. [PMID: 34672299 DOI: 10.21307/ane-2021-027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The present study aimed to examine whether the attenuation of estrogen receptor expression is prevented by propyl pyrazole triol (PPT), an agonist for estrogen receptor α (ERα) or and diarypropiolnitrile (DPN), an agonist for estrogen receptor β (ERβ) after traumatic brain injury (TBI). The tests performed on ovariectomized female Wistar rats included sham group, vehicle group, and treated groups: PPT, DPN, and PPT+DPN 30 minutes after TBI. Blood‑brain barrier (BBB) disruption and brain water content were estimated. RT‑PCR and\r\nwestern blotting were utilized to evaluate ESR1 and ESR2 gene and protein expression. The data indicated that PPT, DPN, and PPT+DPN attenuated TBI‑induced brain edema. Also, BBB disruption after TBI was prevented in PPT, DPN, and PPT+DPN‑treated TBI animals. Estrogen agonist‑treated animals showed a significant elevation in Esr1 mRNA and protein expression levels in the brain tissue of TBI rats. In addition, the data indicated a significant elevation of Esr2 mRNA and protein expression levels in the brain tissue of estrogen agonist‑treated TBI rats. The data shows that both ESR1 and ESR2 agonists can enhance ER mRNA and protein levels in TBI animals' brain. It appears that this effect contributes to the neuroprotective function of ER agonists.
Collapse
Affiliation(s)
- Mohammad Khaksari
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran;
| | - Zahra Hajializadeh
- Endocrinology and Metabolism Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Saeed Esmaeili Mahani
- Department of Biology, Faculty of Sciences, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Zahra Soltani
- Endocrinology and Metabolism Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Gholamreza Asadikaram
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
20
|
Wan L, Huang RJ, Luo ZH, Gong JE, Pan A, Manavis J, Yan XX, Xiao B. Reproduction-Associated Hormones and Adult Hippocampal Neurogenesis. Neural Plast 2021; 2021:3651735. [PMID: 34539776 PMCID: PMC8448607 DOI: 10.1155/2021/3651735] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 08/17/2021] [Indexed: 11/18/2022] Open
Abstract
The levels of reproduction-associated hormones in females, such as estrogen, progesterone, prolactin, and oxytocin, change dramatically during pregnancy and postpartum. Reproduction-associated hormones can affect adult hippocampal neurogenesis (AHN), thereby regulating mothers' behavior after delivery. In this review, we first briefly introduce the overall functional significance of AHN and the methods commonly used to explore this front. Then, we attempt to reconcile the changes of reproduction-associated hormones during pregnancy. We further update the findings on how reproduction-related hormones influence adult hippocampal neurogenesis. This review is aimed at emphasizing a potential role of AHN in reproduction-related brain plasticity and its neurobiological relevance to motherhood behavior.
Collapse
Affiliation(s)
- Lily Wan
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Rou-Jie Huang
- Medical Doctor Program, Xiangya School of Medicine, Central South University, Changsha, China
| | - Zhao-Hui Luo
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Jiao-e Gong
- Department of Neurology, Hunan Children's Hospital, Changsha 410007, China
| | - Aihua Pan
- Department of Anatomy and Neurobiology, Central South University Xiangya School of Medicine, Changsha, Hunan 410013, China
| | - Jim Manavis
- Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, Australia 5000
| | - Xiao-Xin Yan
- Department of Anatomy and Neurobiology, Central South University Xiangya School of Medicine, Changsha, Hunan 410013, China
| | - Bo Xiao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| |
Collapse
|
21
|
Sun Y, Leng P, Guo P, Gao H, Liu Y, Li C, Li Z, Zhang H. G protein coupled estrogen receptor attenuates mechanical stress-mediated apoptosis of chondrocyte in osteoarthritis via suppression of Piezo1. Mol Med 2021; 27:96. [PMID: 34454425 PMCID: PMC8403401 DOI: 10.1186/s10020-021-00360-w] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 08/18/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Apoptosis of chondrocyte is involved in osteoarthritis (OA) pathogenesis, and mechanical stress plays a key role in this process by activation of Piezo1. However, the negative regulation of signal conduction mediated by mechanical stress is still unclear. Here, we elucidate that the critical role of G protein coupled estrogen receptor (GPER) in the regulation of mechanical stress-mediated signal transduction and chondrocyte apoptosis. METHODS The gene expression profile was detected by gene chip upon silencing Piezo1. The expression of GPER in cartilage tissue taken from the clinical patients was detected by RT-PCR and Western blot as well as immunohistochemistry, and the correlation between GPER expression and OA was also investigated. The chondrocytes exposed to mechanical stress were treated with estrogen, G-1, G15, GPER-siRNA and YAP (Yes-associated protein)-siRNA. The cell viability of chondrocytes was measured. The expression of polymerized actin and Piezo1 as well as the subcellular localization of YAP was observed under laser confocal microscope. Western blot confirmed the changes of YAP/ Rho GTPase activating protein 29 (ARHGAP29) /RhoA/LIMK /Cofilin pathway. The knee specimens of osteoarthritis model were stained with safranin and green. OARSI score was used to evaluate the joint lesions. The expressions of GPER and YAP were detected by immunochemistry. RESULTS Expression profiles of Piezo1- silenced chondrocytes showed that GPER expression was significantly upregulated. Moreover, GPER was negatively correlated with cartilage degeneration during OA pathogenesis. In addition, we uncovered that GPER directly targeted YAP and broadly restrained mechanical stress-triggered actin polymerization. Mechanism studies revealed that GPER inhibited mechanical stress-mediated RhoA/LIMK/cofilin pathway, as well as the actin polymerization, by promoting expression of YAP and ARHGAP29, and the YAP nuclear localization, eventually causing the inhibition of Piezo1. YAP was obviously decreased in degenerated cartilage. Silencing YAP caused significantly increased actin polymerization and activation of Piezo1, and an increase of chondrocyte apoptosis. In addition, intra-articular injection of G-1 to OA rat effectively attenuated cartilage degeneration. CONCLUSION We propose a novel regulatory mechanism underlying mechanical stress-mediated apoptosis of chondrocyte and elucidate the potential application value of GPER as therapy targets for OA.
Collapse
Affiliation(s)
- Yi Sun
- Department of Joint Surgery, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Ping Leng
- Department of Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Pengcheng Guo
- Department of Joint Orthopedics, Weifang Hospital of Traditional Chinese Medicine, Weifang, 261000, China
| | - Huanshen Gao
- Department of Joint Surgery, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Yikai Liu
- Department of Joint Surgery, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Chenkai Li
- Department of Joint Surgery, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Zhenghui Li
- Department of Joint Surgery, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Haining Zhang
- Department of Joint Surgery, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China.
| |
Collapse
|
22
|
Batallán Burrowes AA, Sundarakrishnan A, Bouhour C, Chapman CA. G protein-coupled estrogen receptor-1 enhances excitatory synaptic responses in the entorhinal cortex. Hippocampus 2021; 31:1191-1201. [PMID: 34399010 DOI: 10.1002/hipo.23383] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 07/16/2021] [Accepted: 07/24/2021] [Indexed: 12/22/2022]
Abstract
Activation of estrogen receptors is thought to modulate cognitive function in the hippocampus, prefrontal cortex, and striatum by affecting both excitatory and inhibitory synaptic transmission. The entorhinal cortex is a major source of cortical sensory and associational input to the hippocampus, but it is unclear whether either estrogens or progestogens may modulate cognitive function through effects on synaptic transmission in the entorhinal cortex. This study assessed the effects of the brief application of either 17-β estradiol (E2) or progesterone on excitatory glutamatergic synaptic transmission in the female rat entorhinal cortex in vitro. Rats were ovariectomized on postnatal day (PD) 63 and also received subdermal E2 implants to maintain constant low levels of circulating E2 on par with estrus. Electrophysiological recordings from brain slices were obtained between PD70 and PD86, and field excitatory postsynaptic potentials (fEPSPs) reflecting the activation of the superficial layers of the entorhinal cortex were evoked by the stimulation of layer I afferents. The application of E2 (10 nM) for 20 min resulted in a small increase in the amplitude of fEPSPs that reversed during the 30-min washout period. The application of the ERα agonist propylpyrazoletriol (PPT) (100 nM) or the β agonist DPN (1 μM) did not significantly affect synaptic responses. However, the application of the G protein-coupled estrogen receptor-1 (GPER1) agonist G1 (100 nM) induced a reversible increase in fEPSP amplitude similar to that induced by E2. Furthermore, the potentiation of responses induced by G1 was blocked by the GPER1 antagonist G15 (1 μM). Application of progesterone (100 nM) or its metabolite allopregnanolone (1 μM) did not significantly affect synaptic responses. The potentiation of synaptic transmission in the entorhinal cortex induced by the activation of GPER1 receptors may contribute to the modulation of cognitive function in female rats.
Collapse
Affiliation(s)
- Ariel A Batallán Burrowes
- Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, Montréal, Québec, Canada
| | - Adithi Sundarakrishnan
- Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, Montréal, Québec, Canada
| | - Camille Bouhour
- Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, Montréal, Québec, Canada
| | - Clifton Andrew Chapman
- Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, Montréal, Québec, Canada
| |
Collapse
|
23
|
Li X, Johann S, Rune GM, Bender RA. Sex-specific Regulation of Spine Density and Synaptic Proteins by G-protein-coupled Estrogen Receptor (GPER)1 in Developing Hippocampus. Neuroscience 2021; 472:35-50. [PMID: 34364953 DOI: 10.1016/j.neuroscience.2021.07.035] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 07/28/2021] [Accepted: 07/29/2021] [Indexed: 11/19/2022]
Abstract
G-protein-coupled-estrogen-receptor 1 (GPER1) is a membrane-bound receptor that mediates estrogen signaling via intracellular signaling cascades. We recently showed that GPER1 promotes the distal dendritic enrichment of hyperpolarization activated and cyclic nucleotide-gated (HCN)1 channels in CA1 stratum lacunosum-moleculare (SLM), suggesting a role of GPER1-mediated signaling in neuronal plasticity. Here we studied whether this role involves processes of structural plasticity, such as the regulation of spine and synapse density in SLM. In organotypic entorhino-hippocampal cultures from mice expressing eGFP, we analyzed spine densities in SLM after treatment with GPER1 agonist G1 (20 nM). G1 significantly increased the density of "non-stubby" spines (maturing spines with a spine head and a neck), but did so only in cultures from female mice. In support of this finding, the expression of synaptic proteins was sex-specifically altered in the cultures: G1 increased the protein (but not mRNA) expression of PSD95 and reduced the p-/n-cofilin ratio only in cultures from females. Application of E2 (2 nM) reproduced the sex-specific effect on spine density in SLM, but only partially on the expression of synaptic proteins. Spine synapse density was, however, not altered after G1-treatment, suggesting that the increased spine density did not translate into an increased spine synapse density in the culture model. Taken together, our results support a role of GPER1 in mediating structural plasticity in CA1 SLM, but suggest that in developing hippocampus, this role is sex-specific.
Collapse
Affiliation(s)
- Xiaoyu Li
- Institute of Neuroanatomy, University Medical Center Hamburg, 20246 Hamburg, Germany
| | - Sonja Johann
- Institute of Neuroanatomy, University Medical Center Hamburg, 20246 Hamburg, Germany
| | - Gabriele M Rune
- Institute of Neuroanatomy, University Medical Center Hamburg, 20246 Hamburg, Germany
| | - Roland A Bender
- Institute of Neuroanatomy, University Medical Center Hamburg, 20246 Hamburg, Germany.
| |
Collapse
|
24
|
Zhang X, Yang Y, Guo L, Zhou J, Niu J, Wang P, Qiang Y, Liu K, Wen Y, Zhang L, Wang F. GPER1 Modulates Synaptic Plasticity During the Development of Temporal Lobe Epilepsy in Rats. Neurochem Res 2021; 46:2019-2032. [PMID: 34076791 DOI: 10.1007/s11064-021-03336-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 04/23/2021] [Accepted: 04/27/2021] [Indexed: 11/30/2022]
Abstract
G-protein coupled estrogen receptor 1 (GPER1) is a novel type of estrogen receptor. Several studies have shown that it has an anti-inflammatory action,which plays an important role in remyelination and cognitive ability adjustment. However, whether it is involved in the development of temporal lobe epilepsy (TLE) is still unknown. The present study established a TLE model by intraperitoneal injection of lithium chloride (3 mmol/kg) and pilocarpine (50 mg/kg) in rats to study the effect of GPER1 in the synaptic plasticity during the development of temporal lobe epilepsy. A microinjection cannula was implanted into the lateral ventricle region of rats via a stereotaxic instrument. G-1 is the specific GPER1 agonist and G15 is the specific GPER1 antagonist. The G1 or G15 and Dimethyl sulfoxide were injected into the rat brains in the intervention groups and control group, respectively. After G1 intervention, the learning and memory abilities and hippocampal neuron damage in epileptic rats were significantly improved, while G15 weakened the neuroprotective effect of GPER1. Meanwhile, G1 controlled the abnormal formation of hippocampal mossy fiber sprouting caused by seizures, and participated in the regulation of synaptic plasticity by reducing the expression of Synapsin I and increasing the expression of gephyrin. Inhibitory synapse gephyrin may play a significant role in synaptic plasticity.
Collapse
Affiliation(s)
- Xian Zhang
- Ningxia Key Laboratory of Cerebrocranial Diseases, Incubation Base of the National Key Laboratory, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750001, Ningxia, China
| | - Yang Yang
- Ningxia Key Laboratory of Cerebrocranial Diseases, Incubation Base of the National Key Laboratory, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750001, Ningxia, China
- School of Basic Medical Sciences, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750001, Ningxia, China
| | - Li Guo
- School of Basic Medical Sciences, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750001, Ningxia, China
| | - Jinyu Zhou
- School of Public Health and Management, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750001, Ningxia, China
| | - Jianguo Niu
- Ningxia Key Laboratory of Cerebrocranial Diseases, Incubation Base of the National Key Laboratory, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750001, Ningxia, China
- School of Basic Medical Sciences, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750001, Ningxia, China
| | - Peng Wang
- Ningxia Key Laboratory of Cerebrocranial Diseases, Incubation Base of the National Key Laboratory, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750001, Ningxia, China
| | - Yuanyuan Qiang
- Ningxia Key Laboratory of Cerebrocranial Diseases, Incubation Base of the National Key Laboratory, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750001, Ningxia, China
| | - Kunmei Liu
- Ningxia Key Laboratory of Cerebrocranial Diseases, Incubation Base of the National Key Laboratory, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750001, Ningxia, China
| | - Yujun Wen
- Ningxia Key Laboratory of Cerebrocranial Diseases, Incubation Base of the National Key Laboratory, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750001, Ningxia, China
- School of Basic Medical Sciences, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750001, Ningxia, China
| | - Lianxiang Zhang
- Ningxia Key Laboratory of Cerebrocranial Diseases, Incubation Base of the National Key Laboratory, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750001, Ningxia, China.
- School of Basic Medical Sciences, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750001, Ningxia, China.
| | - Feng Wang
- Ningxia Key Laboratory of Cerebrocranial Diseases, Incubation Base of the National Key Laboratory, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750001, Ningxia, China.
- Department of Neurosurgery, General Hospital of Ningxia Medical University, 804 Shengli Street, Yinchuan, 750001, Ningxia, China.
| |
Collapse
|
25
|
Activation of microglial G‑protein-coupled receptor 30 protects neurons against excitotoxicity through NF-κB/MAPK pathways. Brain Res Bull 2021; 172:22-30. [PMID: 33848615 DOI: 10.1016/j.brainresbull.2021.04.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 03/18/2021] [Accepted: 04/07/2021] [Indexed: 11/21/2022]
Abstract
Neuroexcitotoxicity is a common feature in neuronal damage and neurodegenerative diseases. Our previous studies have confirmed that neuronal and astrocytic G‑protein-coupled receptor 30 (GPR30) play a key role in neuroprotection in vivo and in vitro. Microglia are considered as immune cells in the central nervous system. However, the role of microglial GPR30 in neuroprotection against neuroexcitotoxicity remained unclear. In this study, MTT, Western blot, immunocytochemical staining, phagocytosis assay and wound healing assay were employed to detect the effect of GPR30 in N9 microglial cells after exposure to glutamate. We found that the treatment of GPR30 specific agonist G1 inhibited glutamate-induced proliferation and activation in N9 microglial cells. G1 inhibited M1 polarization, facilitated M2 polarization, and decreased over-phagocytosis but had no effect on migration ability in microglia. The result of neurons and microglia co-culture showed that the activation of microglial GPR30 protected neurons from excitotoxicity through the NF-κB/MAPK signaling pathways. Our findings suggested a key role of microglial GPR30 in excitatory neuronal damage and neurodegenerative diseases.
Collapse
|
26
|
Scaplen KM, Petruccelli E. Receptors and Channels Associated with Alcohol Use: Contributions from Drosophila. Neurosci Insights 2021; 16:26331055211007441. [PMID: 33870197 PMCID: PMC8020223 DOI: 10.1177/26331055211007441] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 03/03/2021] [Indexed: 12/14/2022] Open
Abstract
Alcohol Use Disorder (AUD) is a debilitating disorder that manifests as problematic patterns of alcohol use. At the core of AUD's behavioral manifestations are the profound structural, physiological, cellular, and molecular effects of alcohol on the brain. While the field has made considerable progress in understanding the neuromolecular targets of alcohol we still lack a comprehensive understanding of alcohol's actions and effective treatment strategies. Drosophila melanogaster is a powerful model for investigating the neuromolecular targets of alcohol because flies model many of the core behavioral elements of AUD and offer a rich genetic toolkit to precisely reveal the in vivo molecular actions of alcohol. In this review, we focus on receptors and channels that are often targeted by alcohol within the brain. We discuss the general roles of these proteins, their role in alcohol-associated behaviors across species, and propose ways in which Drosophila models can help advance the field.
Collapse
Affiliation(s)
- Kristin M Scaplen
- Department of Psychology, Bryant University, Smithfield, RI, USA
- Center for Health and Behavioral Studies, Bryant University, Smithfield, RI, USA
- Department of Neuroscience, Brown University, Providence, RI, USA
| | - Emily Petruccelli
- Department of Biological Sciences, Southern Illinois University Edwardsville, Edwardsville, IL, USA
| |
Collapse
|
27
|
Chou HT, Wu PY, Huang JC, Chen SC, Ho WY. Late Menarche, Not Reproductive Period, Is Associated with Poor Cognitive Function in Postmenopausal Women in Taiwan. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:2345. [PMID: 33673620 PMCID: PMC7967768 DOI: 10.3390/ijerph18052345] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 02/17/2021] [Accepted: 02/23/2021] [Indexed: 01/09/2023]
Abstract
Female sex hormones such as estrogen and progesterone play an important role in the regulation of a woman's body, including cognition and neurogenesis. However, the effects of age at menarche and reproductive period on cognitive function are still controversial. The aim of this study was to investigate the relationships between age at menarche and reproductive period with cognitive impairment. Data were obtained from the Taiwan Biobank. Cognitive function was assessed using the Mini Mental State Examination (MMSE) and its five subdomains. Multivariable linear regression analysis revealed that an old age at menarche (per one year; coefficient β, -0.189; p = 0.020) was significantly associated with a low total MMSE score, whereas reproductive period (p = 0.733) was not significantly associated with total MMSE score. Furthermore, an old age at menarche was significantly associated with low MMSE G2 (registration) (per one year; coefficient β, -0.022; p = 0.035) and G5 (language, construction and obey) scores (per one year; coefficient β, -0.054; p = 0.047). However, age at menarche was not significantly associated with MMSE G1 (orientation), G3 (attention and calculation) and G4 (recall) scores. In addition, reproductive period was not significantly associated with any MMSE subscores. Late menarche was associated with poor cognitive function, including low total MMSE score and low MMSE G2 and G5 scores. However, reproductive period was not associated with cognitive function in postmenopausal women.
Collapse
Affiliation(s)
- Hung-Tse Chou
- Department of General Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan;
| | - Pei-Yu Wu
- Department of Internal Medicine, Kaohsiung Municipal Siaogang Hospital, Kaohsiung Medical University, Kaohsiung 812, Taiwan; (P.-Y.W.); (J.-C.H.); (S.-C.C.)
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Jiun-Chi Huang
- Department of Internal Medicine, Kaohsiung Municipal Siaogang Hospital, Kaohsiung Medical University, Kaohsiung 812, Taiwan; (P.-Y.W.); (J.-C.H.); (S.-C.C.)
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Szu-Chia Chen
- Department of Internal Medicine, Kaohsiung Municipal Siaogang Hospital, Kaohsiung Medical University, Kaohsiung 812, Taiwan; (P.-Y.W.); (J.-C.H.); (S.-C.C.)
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Research Center for Environmental Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Wan-Yi Ho
- Department of Anatomy, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| |
Collapse
|
28
|
Ma HY, Chen S, Du Y. Estrogen and estrogen receptors in kidney diseases. Ren Fail 2021; 43:619-642. [PMID: 33784950 PMCID: PMC8018493 DOI: 10.1080/0886022x.2021.1901739] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 03/03/2021] [Accepted: 03/06/2021] [Indexed: 02/08/2023] Open
Abstract
Acute kidney injury (AKI) and chronic kidney disease (CKD) are posing great threats to global health within this century. Studies have suggested that estrogen and estrogen receptors (ERs) play important roles in many physiological processes in the kidney. For instance, they are crucial in maintaining mitochondrial homeostasis and modulating endothelin-1 (ET-1) system in the kidney. Estrogen takes part in the kidney repair and regeneration via its receptors. Estrogen also participates in the regulation of phosphorus homeostasis via its receptors in the proximal tubule. The ERα polymorphisms have been associated with the susceptibilities and outcomes of several renal diseases. As a consequence, the altered or dysregulated estrogen/ERs signaling pathways may contribute to a variety of kidney diseases, including various causes-induced AKI, diabetic kidney disease (DKD), lupus nephritis (LN), IgA nephropathy (IgAN), CKD complications, etc. Experimental and clinical studies have shown that targeting estrogen/ERs signaling pathways might have protective effects against certain renal disorders. However, many unsolved problems still exist in knowledge regarding the roles of estrogen and ERs in distinct kidney diseases. Further research is needed to shed light on this area and to enable the discovery of pathway-specific therapies for kidney diseases.
Collapse
Affiliation(s)
- Hao-Yang Ma
- Department of Geriatrics, Second Affiliated Hospital of Zhejiang University, Hangzhou, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Shuang Chen
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Yang Du
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
29
|
Ambrase A, Lewis CA, Barth C, Derntl B. Influence of ovarian hormones on value-based decision-making systems: Contribution to sexual dimorphisms in mental disorders. Front Neuroendocrinol 2021; 60:100873. [PMID: 32987043 DOI: 10.1016/j.yfrne.2020.100873] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 07/28/2020] [Accepted: 09/15/2020] [Indexed: 12/22/2022]
Abstract
Women and men exhibit differences in behavior when making value-based decisions. Various hypotheses have been proposed to explain these findings, stressing differences in functional lateralization of the brain, functional activation, neurotransmitter involvement and more recently, sex hormones. While a significant interaction of neurotransmitter systems and sex hormones has been shown for both sexes, decision-making in women might be particularly affected by variations of ovarian hormones. In this review we have gathered information from animal and human studies on how ovarian hormones affect decision-making processes in females by interacting with neurotransmitter systems at functionally relevant brain locations and thus modify the computation of decision aspects. We also review previous findings on impaired decision-making in animals and clinical populations with substance use disorder and depression, emphasizing how little we know about the role of ovarian hormones in aberrant decision-making.
Collapse
Affiliation(s)
- Aiste Ambrase
- Department of Psychiatry and Psychotherapy, University of Tuebingen, Tübingen, Germany; International Max Planck Research School for Cognitive and Systems Neuroscience, University of Tübingen, Tuebingen, Germany
| | - Carolin A Lewis
- Department of Psychiatry and Psychotherapy, University of Tuebingen, Tübingen, Germany; Emotion Neuroimaging Lab, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany; International Max Planck Research School on Neuroscience of Communication: Function, Structure, and Plasticity, Leipzig, Germany
| | - Claudia Barth
- Norwegian Centre for Mental Disorders Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Birgit Derntl
- Department of Psychiatry and Psychotherapy, University of Tuebingen, Tübingen, Germany; International Max Planck Research School for Cognitive and Systems Neuroscience, University of Tübingen, Tuebingen, Germany; TübingenNeuroCampus, University of Tübingen, Tübingen, Germany; LEAD Research School and Graduate Network, University of Tübingen, Tübingen, Germany.
| |
Collapse
|
30
|
Ariyani W, Miyazaki W, Amano I, Hanamura K, Shirao T, Koibuchi N. Soy Isoflavones Accelerate Glial Cell Migration via GPER-Mediated Signal Transduction Pathway. Front Endocrinol (Lausanne) 2020; 11:554941. [PMID: 33250856 PMCID: PMC7672195 DOI: 10.3389/fendo.2020.554941] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 10/12/2020] [Indexed: 01/04/2023] Open
Abstract
Soybean isoflavones, such as genistein, daidzein, and its metabolite, S-equol, are widely known as phytoestrogens. Their biological actions are thought to be exerted via the estrogen signal transduction pathway. Estrogens, such as 17β-estradiol (E2), play a crucial role in the development and functional maintenance of the central nervous system. E2 bind to the nuclear estrogen receptor (ER) and regulates morphogenesis, migration, functional maturation, and intracellular metabolism of neurons and glial cells. In addition to binding to nuclear ER, E2 also binds to the G-protein-coupled estrogen receptor (GPER) and activates the nongenomic estrogen signaling pathway. Soybean isoflavones also bind to the ER and GPER. However, the effect of soybean isoflavone on brain development, particularly glial cell function, remains unclear. We examined the effects of soybean isoflavones using an astrocyte-enriched culture and astrocyte-derived C6 clonal cells. Isoflavones increased glial cell migration. This augmentation was suppressed by co-exposure with G15, a selective GPER antagonist, or knockdown of GPER expression using RNA interference. Isoflavones also activated actin cytoskeleton arrangement via increased actin polymerization and cortical actin, resulting in an increased number and length of filopodia. Isoflavones exposure increased the phosphorylation levels of FAK (Tyr397 and Tyr576/577), ERK1/2 (Thr202/Tyr204), Akt (Ser473), and Rac1/cdc42 (Ser71), and the expression levels of cortactin, paxillin and ERα. These effects were suppressed by knockdown of the GPER. Co-exposure of isoflavones to the selective RhoA inhibitor, rhosin, selective Cdc42 inhibitor, casin, or Rac1/Cdc42 inhibitor, ML-141, decreased the effects of isoflavones on cell migration. These findings indicate that soybean isoflavones exert their action via the GPER to activate the PI3K/FAK/Akt/RhoA/Rac1/Cdc42 signaling pathway, resulting in increased glial cell migration. Furthermore, in silico molecular docking studies to examine the binding mode of isoflavones to the GPER revealed the possibility that isoflavones bind directly to the GPER at the same position as E2, further confirming that the effects of the isoflavones are at least in part exerted via the GPER signal transduction pathway. The findings of the present study indicate that isoflavones may be an effective supplement to promote astrocyte migration in developing and/or injured adult brains.
Collapse
Affiliation(s)
- Winda Ariyani
- Research Fellow of Japan Society for the Promotion of Science, Tokyo, Japan
- Department of Integrative Physiology, Graduate School of Medicine, Gunma University, Maebashi, Japan
| | - Wataru Miyazaki
- Department of Integrative Physiology, Graduate School of Medicine, Gunma University, Maebashi, Japan
- Department of Bioscience and Laboratory Medicine, Graduate School of Health Science, Hirosaki University, Hirosaki, Japan
| | - Izuki Amano
- Department of Integrative Physiology, Graduate School of Medicine, Gunma University, Maebashi, Japan
| | - Kenji Hanamura
- Department of Neurobiology and Behavior, Graduate School of Medicine, Gunma University, Maebashi, Japan
| | - Tomoaki Shirao
- Department of Neurobiology and Behavior, Graduate School of Medicine, Gunma University, Maebashi, Japan
| | - Noriyuki Koibuchi
- Department of Integrative Physiology, Graduate School of Medicine, Gunma University, Maebashi, Japan
| |
Collapse
|
31
|
McCarthy M, Raval AP. The peri-menopause in a woman's life: a systemic inflammatory phase that enables later neurodegenerative disease. J Neuroinflammation 2020; 17:317. [PMID: 33097048 PMCID: PMC7585188 DOI: 10.1186/s12974-020-01998-9] [Citation(s) in RCA: 113] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 10/14/2020] [Indexed: 02/08/2023] Open
Abstract
The peri-menopause or menopausal transition—the time period that surrounds the final years of a woman’s reproductive life—is associated with profound reproductive and hormonal changes in a woman’s body and exponentially increases a woman’s risk of cerebral ischemia and Alzheimer’s disease. Although our understanding of the exact timeline or definition of peri-menopause is limited, it is clear that there are two stages to the peri-menopause. These are the early menopausal transition, where menstrual cycles are mostly regular, with relatively few interruptions, and the late transition, where amenorrhea becomes more prolonged and lasts for at least 60 days, up to the final menstrual period. Emerging evidence is showing that peri-menopause is pro-inflammatory and disrupts estrogen-regulated neurological systems. Estrogen is a master regulator that functions through a network of estrogen receptors subtypes alpha (ER-α) and beta (ER-β). Estrogen receptor-beta has been shown to regulate a key component of the innate immune response known as the inflammasome, and it also is involved in regulation of neuronal mitochondrial function. This review will present an overview of the menopausal transition as an inflammatory event, with associated systemic and central nervous system inflammation, plus regulation of the innate immune response by ER-β-mediated mechanisms.
Collapse
Affiliation(s)
- Micheline McCarthy
- Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, 33136, USA
| | - Ami P Raval
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratory, Leonard M. Miller School of Medicine, University of Miami, 1420 NW 9th Avenue, Neurology Research Building, Room # 203H, Miami, FL, 33136, USA. .,Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, 33136, USA.
| |
Collapse
|
32
|
Amirkhosravi L, Khaksari M, Soltani Z, Esmaeili-Mahani S, Asadi Karam G, Hoseini M. E2-BSA and G1 exert neuroprotective effects and improve behavioral abnormalities following traumatic brain injury: The role of classic and non-classic estrogen receptors. Brain Res 2020; 1750:147168. [PMID: 33096091 DOI: 10.1016/j.brainres.2020.147168] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/30/2020] [Accepted: 10/15/2020] [Indexed: 12/15/2022]
Abstract
The role of classical and non-classical estrogen receptors (ERs) in mediating the neuroprotective effects of this hormone on brain edema and long-term behavioral disorders was evaluated after traumatic brain injury (TBI). Ovariectomized rats were divided as follows: E2 (17 β-estradiol), E2-BSA (E2 conjugated to bovine serum albumin), G1 [G-protein-coupled estrogen receptor agonist (GPER)] or their vehicle was injected following TBI, whereas ICI (classical estrogen receptor antagonist), G15 (GPER antagonist), ICI + G15, and their vehicle were injected before the induction of TBI and the injection of E2 and E2-BSA. Brain water (BWC) and Evans blue (EB) contents were measured 24 h and 5 h after TBI, respectively. Intracranial pressure (ICP) and cerebral perfusion pressure (CPP) were measured before and at different times after TBI. Locomotor activity, anxiety-like behavior, and spatial memory were assessed on days 3, 7, 14, and 21 after injury. E2, E2-BSA, and G1 prevented the increase of BWC and EB content after TBI, and these effects were inhibited by ICI and G15. ICI and G15 also inhibited the beneficial effects of E2, E2-BSA on ICP, as well as CPP, after trauma. E2, E2-BSA, and G1 prevented the cognitive deficiency and behavioral abnormalities induced by TBI. Similar to the above parameters, ICI and G15 also reversed this E2 and E2-BSA effects on days 3, 7, 14, and 21. Our findings indicated that the beneficial effects of E2-BSA and E2 were inhibited by both ICI and G15, suggesting that GPER and classic ERs were involved in mediating the long-term effects of E2.
Collapse
Affiliation(s)
- Ladan Amirkhosravi
- Department of Physiology, Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran; Endocrinology and Metabolism Research Center, Institute of Basic and Clinical Physiology Sciences, Faculty of Medicine, Kerman, Iran
| | - Mohammad Khaksari
- Neuroscience and Endocrinology and Metabolism Research Centers, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran.
| | - Zahra Soltani
- Endocrinology and Metabolism Research Center, Institute of Basic and Clinical Physiology Sciences, Faculty of Medicine, Kerman, Iran
| | - Saeed Esmaeili-Mahani
- Department of Biology, Faculty of Sciences, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Gholamreza Asadi Karam
- Department of Biochemistry, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Mojtaba Hoseini
- Department of Biology, Faculty of Sciences, Shahid Bahonar University of Kerman, Kerman, Iran
| |
Collapse
|
33
|
Acosta-Martínez M. Shaping Microglial Phenotypes Through Estrogen Receptors: Relevance to Sex-Specific Neuroinflammatory Responses to Brain Injury and Disease. J Pharmacol Exp Ther 2020; 375:223-236. [PMID: 32513838 DOI: 10.1124/jpet.119.264598] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 05/05/2020] [Indexed: 03/08/2025] Open
Abstract
In mammals, 17β-estradiol (E2), the primary endogenous estrogen, maintains normal central nervous system (CNS) function throughout life and influences brain responses to injury and disease. Estradiol-induced cellular changes are mediated through the activation of nuclear and extranuclear estrogen receptors (ERs), which include ERα, ERβ, and the G-protein coupled receptor, GPER1. ERs are widely expressed throughout the brain, acting as transcriptional effectors or rapidly initiating membrane and cytoplasmic signaling cascades in practically all brain cells including microglia, the resident immune cells of the CNS. Activation of ERs by E2 exerts potent anti-inflammatory effects through mechanisms involving the modification of microglial cell responses to acute or chronic brain injury. Recent studies suggest that microglial maturation is influenced by the internal gonadal hormone milieu and that their functions in the normal and diseased brain are sex specific. However, the role that each ER subtype plays in microglial development or in determining microglial function as the primary cellular defense mechanism against pathogens and injury remains unclear. This is partly due to the fact that most studies investigating the mechanisms by which E2-ER signaling modifies microglial cellular phenotypes have been restricted to one sex or age. This review examines the different in vivo and in vitro models used to study the direct and indirect regulation of microglial cell function by E2 through ERs. Ischemic stroke will be used as an example of a neurologic disease in which activation of ERs shapes microglial phenotypes in response to injury in a sex- and age-specific fashion. SIGNIFICANCE STATEMENT: As the primary immune sensors of central nervous system damage, microglia are important potential therapeutic targets. Estrogen receptor signaling modulates microglial responses to brain injury and disease in a sex- and age-specific fashion. Hence, investigating the molecular mechanisms by which estrogen receptors regulate and shape microglial functions throughout life may result in novel and effective therapeutic opportunities that are tailored for each sex and age.
Collapse
|
34
|
Kirshner ZZ, Yao JK, Li J, Long T, Nelson D, Gibbs RB. Impact of estrogen receptor agonists and model of menopause on enzymes involved in brain metabolism, acetyl-CoA production and cholinergic function. Life Sci 2020; 256:117975. [PMID: 32565251 PMCID: PMC7448522 DOI: 10.1016/j.lfs.2020.117975] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 06/11/2020] [Accepted: 06/12/2020] [Indexed: 12/18/2022]
Abstract
Our goal is to understand how loss of circulating estrogens and estrogen replacement affect brain physiology and function, particularly in brain regions involved in cognitive processes. We recently conducted a large metabolomics study characterizing the effects of rodent models of menopause and treatment with estrogen receptor (ER) agonists on neurochemical targets in hippocampus, frontal cortex, and striatum. Here we characterize effects on levels of several key enzymes involved in glucose utilization and energy production, specifically phosphofructokinase, glyceraldehyde 3-phosphate dehydrogenase, and pyruvate dehydrogenase. We also evaluated effects on levels of β-actin and α-tubulin, choline acetyltransferase (ChAT) activity, and levels of ATP citrate lyase. All experiments were conducted in young adult rats. Experiment 1 compared the effects of ovariectomy (OVX), a model of surgical menopause, and 4-vinylcyclohexene diepoxide (VCD)-treatments, a model of transitional menopause, with tissues collected at proestrus and at diestrus. Experiment 2 used a separate cohort of rats to evaluate the same targets in OVX and VCD-treated rats treated with estradiol or with selective ER agonists. Differences in the expression of metabolic enzymes between cycling animals and models of surgical and transitional menopause were detected. These differences were model-, region- and time- dependent, and were modulated by selective ER agonists. Collectively, the findings demonstrate that loss of ovarian function and ER agonist treatments have differing effects in OVX vs. VCD-treated rats. Differences may help to explain differences in the effects of estrogen treatments on brain function and cognition in women who have experienced surgical vs. transitional menopause.
Collapse
Affiliation(s)
- Z Z Kirshner
- University of Pittsburgh, Department of Pharmaceutical Sciences, 1004 Salk Hall, Pittsburgh, PA 15261, USA.
| | - Jeffrey K Yao
- University of Pittsburgh, Department of Pharmaceutical Sciences, 1004 Salk Hall, Pittsburgh, PA 15261, USA.
| | - Junyi Li
- University of Pittsburgh, Department of Pharmaceutical Sciences, 1004 Salk Hall, Pittsburgh, PA 15261, USA.
| | - Tao Long
- University of Pittsburgh, Department of Pharmaceutical Sciences, 1004 Salk Hall, Pittsburgh, PA 15261, USA.
| | - Doug Nelson
- University of Pittsburgh, Department of Pharmaceutical Sciences, 1004 Salk Hall, Pittsburgh, PA 15261, USA.
| | - R B Gibbs
- University of Pittsburgh, Department of Pharmaceutical Sciences, 1004 Salk Hall, Pittsburgh, PA 15261, USA.
| |
Collapse
|
35
|
Sex Differences in Lower Limb Proprioception and Mechanical Function Among Healthy Adults. Motor Control 2020; 24:571-587. [PMID: 32866946 DOI: 10.1123/mc.2020-0015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 06/01/2020] [Accepted: 06/05/2020] [Indexed: 11/18/2022]
Abstract
Twenty-four healthy adults, including 12 females and 12 males, participated in the study. Each female participant completed three trials in three different phases of one menstrual cycle, which included follicular, ovulatory, and luteal phases. The study aimed to investigate whether there is any difference in joint kinetic sense, neuromuscular coordination, and isokinetic muscle strength (a) between healthy males and females at different phases of the menstrual cycle and (b) between females at different phases of the menstrual cycle. The outcome measures included the number of jumps in the square-hop test and ankle and knee proprioception, which were assessed by an electric-driven movable frame rotated at 0.4 deg/s and isokinetic muscle strength measured by a computerized dynamometer (Biodex). For the square-hop test (p = .006), ankle dorsiflexion/plantar flexion (p < .05), knee flexion/extension (p < .05), the relative peak torque of the isokinetic muscle strength at the 60° and 180° knee flexion/extension (p < .001), and the 30° and 120° ankle plantar flexion/dorsiflexion (p < .05) between females and males showed significant differences. For the females at different phases of the menstrual cycle, significant differences were found on ankle dorsiflexion (p = .003), plantar flexion (p = .023), knee extension (p = .029), the square-hop test (p = .036), and relative peak torque of isokinetic muscle strength at 180° knee flexion (p = .029). This study demonstrated that there are sex differences in lower limb proprioception and mechanical function. Females at ovulatory and luteal phases have better lower limb proprioception than at the follicular phase.
Collapse
|
36
|
Non-genomic mechanisms mediate androgen-induced PSD95 expression. Aging (Albany NY) 2020; 11:2281-2294. [PMID: 31005955 PMCID: PMC6520003 DOI: 10.18632/aging.101913] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 04/10/2019] [Indexed: 12/14/2022]
Abstract
The non-genomic actions of androgen-induced synaptic plasticity have been extensively studied. However, the underlying mechanisms remain controversial. We recently found that testosterone-fetal bovine serum albumin (T-BSA), a cell membrane-impermeable complex, led to a rapid increase in the postsynaptic density 95 (PSD95) protein level through a transcription-independent mechanism in mouse hippocampal HT22 cells. Using T-BSA conjugated FITC, we verified the presence of membrane androgen-binding sites. Here, we show that T-BSA-induced PSD95 expression is mediated by G-protein-coupled receptor (GPCR)-zinc transporter ZIP9 (SLC39A9), one of the androgen membrane binding sites, rather than the membrane-localized androgen receptor. Furthermore, we found that T-BSA induced an interaction between ZIP9 and Gnα11 that lead to the phosphorylation of Erk1/2 MAPK and eIF4E, which are critical in the mRNA translation process. The PSD95 and p-eIF4E expression decreased when knockdown of ZIP9 or Gnα11 expression or inhibition of Erk1/2 activation. Taken together, these findings suggest that ZIP9 mediates the non-genomic action of androgen on synaptic protein PSD95 synthesis through the Gnα11/Erk1/2/eIF4E pathway in HT22 cells. This novel mechanism provides a theoretical basis to understand the neuroprotective mechanism of androgen.
Collapse
|
37
|
Naderi M, Salahinejad A, Attaran A, Niyogi S, Chivers DP. Rapid effects of estradiol and its receptor agonists on object recognition and object placement in adult male zebrafish. Behav Brain Res 2020; 384:112514. [PMID: 32004591 DOI: 10.1016/j.bbr.2020.112514] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 01/15/2020] [Accepted: 01/24/2020] [Indexed: 10/25/2022]
Abstract
In recent years, there has been a growing appreciation that 17β-estradiol (E2) can rapidly modulate learning and memory processes by binding to membrane estrogen receptors and cause the activation of a number of signaling cascades within the central nervous system. In this study, we sought to investigate the effects of post-training administration of E2 (100 ng/g, 1 μg/g, 10 μg/g) and involvement of the estrogen receptors (ERs) using selective ER agonists on the consolidation of object recognition (OR) and object placement memory (OP) in adult male zebrafish. The general activation of ERs with the highest E2 dose improved consolidation of memory in both learning tasks within 1.45 h of administration. Activation of classical ERs (ERα and ERβ) improved consolidation of OR memory, but had no effect on fish performance in OP task. On the other hand, activation of G protein-coupled ER1 impaired and enhanced consolidation of OR and OP memories, respectively. Memory improvement in both tasks was accompanied by a marked up-regulation in the expression of genes encoding ionotropic and metabotropic glutamate receptors in a task-dependent manner. In contrast, the down-regulation in the expression of certain ionotropic glutamate receptors was observed in fish with impaired OR memory. Moreover, our study also revealed an increase in the transcript abundance of genes associated with synaptic protein synthesis (brain-derived neurotrophic factor, synaptophysin, and the mechanistic target of rapamycin). These results suggest that E2 may affect consolidation of memory in zebrafish likely through rapid changes in synaptic morphology and function.
Collapse
Affiliation(s)
- Mohammad Naderi
- Department of Biology, University of Saskatchewan, 112 Science Place, Saskatoon, SK S7N 5E2, Canada.
| | - Arash Salahinejad
- Department of Biology, University of Saskatchewan, 112 Science Place, Saskatoon, SK S7N 5E2, Canada
| | - Anoosha Attaran
- Department of Biology, University of Saskatchewan, 112 Science Place, Saskatoon, SK S7N 5E2, Canada
| | - Som Niyogi
- Department of Biology, University of Saskatchewan, 112 Science Place, Saskatoon, SK S7N 5E2, Canada; Toxicology Centre, University of Saskatchewan, 44 Campus Drive, Saskatoon, SK S7N 5B3, Canada
| | - Douglas P Chivers
- Department of Biology, University of Saskatchewan, 112 Science Place, Saskatoon, SK S7N 5E2, Canada
| |
Collapse
|
38
|
Zuo D, Wang F, Rong W, Wen Y, Sun K, Zhao X, Ren X, He Z, Ding N, Ma L, Xu F. The novel estrogen receptor GPER1 decreases epilepsy severity and susceptivity in the hippocampus after status epilepticus. Neurosci Lett 2020; 728:134978. [PMID: 32302699 DOI: 10.1016/j.neulet.2020.134978] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 04/04/2020] [Accepted: 04/07/2020] [Indexed: 12/17/2022]
Abstract
The steroid hormone 17β-estradiol (estrogen) exerts neuroprotective effects in several types of neurological disorders including epilepsy. The novel G protein-coupled estrogen receptor 1 (GPER1), also called GPR30, mediates the non-genomic effects of 17β-estradiol. However, the specific role of GPER1 in status epilepticus (SE) remains unclear. In this report, we evaluated the effects of GPER1 on the hippocampus during SE and the underlying mechanism was studied. Our results revealed that pilocarpine-induced GPER1-KD epileptic rats exhibited a shorter latency to generalized convulsions and strikingly elevated seizure severity. Additionally, the electroencephalographic seizure activity also corresponded to these results. Fast-Fourier analysis indicated an enhancement of power in the theta and alpha bands during SE in GPER1-KD rats. In addition, epilepsy-induced pathological changes were dramatically exacerbated in GPER1-KD rats, including neuron damage and neuroinflammation in hippocampus. GPER1 might be associated with the susceptibility to and severity of epileptic seizures. In summary, our results suggested that GPER1 plays a neuroprotective role in SE, and might be a candidate target for epilepsy therapy.
Collapse
Affiliation(s)
- Di Zuo
- Ningxia Key Laboratory of Cerebrocranial Diseases, Incubation Base of the National Key Laboratory, Ningxia Medical University, 1160 Shengli Street, Yinchuan, Ningxia 750001, China; School of Basic Medical Sciences, Ningxia Medical University, 1160 Shengli Street, Yinchuan, Ningxia 750001, China
| | - Feng Wang
- Ningxia Key Laboratory of Cerebrocranial Diseases, Incubation Base of the National Key Laboratory, Ningxia Medical University, 1160 Shengli Street, Yinchuan, Ningxia 750001, China; Department of Neurosurgery, General Hospital of Ningxia Medical University, 804 Shengli Street, Yinchuan, Ningxia 750001, China
| | - Weifang Rong
- School of Basic Medical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yujun Wen
- Ningxia Key Laboratory of Cerebrocranial Diseases, Incubation Base of the National Key Laboratory, Ningxia Medical University, 1160 Shengli Street, Yinchuan, Ningxia 750001, China
| | - Kuisheng Sun
- Ningxia Key Laboratory of Cerebrocranial Diseases, Incubation Base of the National Key Laboratory, Ningxia Medical University, 1160 Shengli Street, Yinchuan, Ningxia 750001, China; Department of Neurosurgery, General Hospital of Ningxia Medical University, 804 Shengli Street, Yinchuan, Ningxia 750001, China
| | - Xiaopeng Zhao
- School of Basic Medical Sciences, Ningxia Medical University, 1160 Shengli Street, Yinchuan, Ningxia 750001, China
| | - Xiaofan Ren
- Ningxia Key Laboratory of Cerebrocranial Diseases, Incubation Base of the National Key Laboratory, Ningxia Medical University, 1160 Shengli Street, Yinchuan, Ningxia 750001, China
| | - Zhenquan He
- Ningxia Key Laboratory of Cerebrocranial Diseases, Incubation Base of the National Key Laboratory, Ningxia Medical University, 1160 Shengli Street, Yinchuan, Ningxia 750001, China
| | - Na Ding
- Ningxia Key Laboratory of Cerebrocranial Diseases, Incubation Base of the National Key Laboratory, Ningxia Medical University, 1160 Shengli Street, Yinchuan, Ningxia 750001, China
| | - Lin Ma
- Ningxia Key Laboratory of Cerebrocranial Diseases, Incubation Base of the National Key Laboratory, Ningxia Medical University, 1160 Shengli Street, Yinchuan, Ningxia 750001, China
| | - Fang Xu
- School of Basic Medical Sciences, Ningxia Medical University, 1160 Shengli Street, Yinchuan, Ningxia 750001, China.
| |
Collapse
|
39
|
Dombret C, Naulé L, Trouillet AC, Parmentier C, Hardin-Pouzet H, Mhaouty-Kodja S. Effects of neural estrogen receptor beta deletion on social and mood-related behaviors and underlying mechanisms in male mice. Sci Rep 2020; 10:6242. [PMID: 32277160 PMCID: PMC7148327 DOI: 10.1038/s41598-020-63427-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 03/30/2020] [Indexed: 12/11/2022] Open
Abstract
Estradiol derived from neural aromatization of testosterone plays a key role in the organization and activation of neural structures underlying male behaviors. This study evaluated the contribution of the estrogen receptor (ER) β in estradiol-induced modulation of social and mood-related behaviors by using mice lacking the ERβ gene in the nervous system. Mutant males exhibited reduced social interaction with same-sex congeners and impaired aggressive behavior. They also displayed increased locomotor activity, and reduced or unaffected anxiety-state level in three paradigms. However, when mice were exposed to unescapable stress in the forced swim and tail suspension tests, they spent more time immobile and a reduced time in swimming and climbing. These behavioral alterations were associated with unaffected circadian and restraint stress-induced corticosterone levels, and unchanged number of tryptophan hydroxylase 2-immunoreactive neurons in the dorsal raphe. By contrast, reduced mRNA levels of oxytocin and arginine-vasopressin were observed in the bed nucleus of stria terminalis, whereas no changes were detected in the hypothalamic paraventricular nucleus. The neural ERβ is thus involved to different extent levels in social and mood-related behaviors, with a particular action on oxytocin and arginine-vasopressin signaling pathways of the bed nucleus of stria terminalis, yet the involvement of other brain areas cannot be excluded.
Collapse
Affiliation(s)
- Carlos Dombret
- Sorbonne Université, CNRS, INSERM, Neuroscience Paris Seine - Institut de Biologie Paris Seine, 75005, Paris, France
| | - Lydie Naulé
- Sorbonne Université, CNRS, INSERM, Neuroscience Paris Seine - Institut de Biologie Paris Seine, 75005, Paris, France
| | - Anne-Charlotte Trouillet
- Sorbonne Université, CNRS, INSERM, Neuroscience Paris Seine - Institut de Biologie Paris Seine, 75005, Paris, France
| | - Caroline Parmentier
- Sorbonne Université, CNRS, INSERM, Neuroscience Paris Seine - Institut de Biologie Paris Seine, 75005, Paris, France
| | - Hélène Hardin-Pouzet
- Sorbonne Université, CNRS, INSERM, Neuroscience Paris Seine - Institut de Biologie Paris Seine, 75005, Paris, France
| | - Sakina Mhaouty-Kodja
- Sorbonne Université, CNRS, INSERM, Neuroscience Paris Seine - Institut de Biologie Paris Seine, 75005, Paris, France.
| |
Collapse
|
40
|
Sarchielli E, Guarnieri G, Idrizaj E, Squecco R, Mello T, Comeglio P, Gallina P, Maggi M, Vannelli GB, Morelli A. The G protein-coupled oestrogen receptor, GPER1, mediates direct anti-inflammatory effects of oestrogens in human cholinergic neurones from the nucleus basalis of Meynert. J Neuroendocrinol 2020; 32:e12837. [PMID: 32077170 DOI: 10.1111/jne.12837] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Revised: 12/18/2019] [Accepted: 01/26/2020] [Indexed: 12/28/2022]
Abstract
It has been well established, particularly in animal models, that oestrogens exert neuroprotective effects in brain areas linked to cognitive processes. A key protective role could reside in the capacity of oestrogen to modulate the inflammatory response. However, the direct neuroprotective actions of oestrogens on neurones are complex and remain to be fully clarified. In the present study, we took advantage of a previously characterised primary culture of human cholinergic neurones (hfNBM) from the foetal nucleus basalis of Meynert, which is known to regulate hippocampal and neocortical learning and memory circuits, aiming to investigate the direct effects of oestrogens under inflammatory conditions. Exposure of cells to tumour necrosis factor (TNF)α (10 ng mL-1 ) determined the activation of an inflammatory response, as demonstrated by nuclear factor-kappa B p65 nuclear translocation and cyclooxygenase-2 mRNA expression. These effects were inhibited by treatment with either 17β-oestradiol (E2 ) (10 nmol L-1 ) or G1 (100 nmol L-1 ), the selective agonist of the G protein-coupled oestrogen receptor (GPER1). Interestingly, the GPER1 antagonist G15 abolished the effects of E2 in TNFα-treated cells, whereas the ERα/ERβ inhibitor tamoxifen did not. Electrophysiological measurements in hfNBMs revealed a depolarising effect caused by E2 that was specifically blocked by tamoxifen and not by G15. Conversely, G1 specifically hyperpolarised the cell membrane and also increased both inward and outward currents elicited by a depolarising stimulus, suggesting a modulatory action on hfNBM excitability by GPER1 activation. Interestingly, pretreating cells with TNFα completely blocked the effects of G1 on membrane properties and also significantly reduced GPER1 mRNA expression. In addition, we found a peculiar subcellular localisation of GPER1 to focal adhesion sites that implicates new possible mechanisms of action of GPER1 in the neuronal perception of mechanical stimuli. The results obtained in the present study indicate a modulatory functional role of GPER1 with respect to mediating the oestrogen neuroprotective effect against inflammation in brain cholinergic neurones and, accordingly, may help to identify protective strategies for preventing cognitive impairments.
Collapse
Affiliation(s)
- Erica Sarchielli
- Section of Human Anatomy and Histology, Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Giulia Guarnieri
- Section of Human Anatomy and Histology, Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Eglantina Idrizaj
- Section of Physiological Sciences, Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Roberta Squecco
- Section of Physiological Sciences, Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Tommaso Mello
- Clinical Gastroenterology Unit, Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Paolo Comeglio
- Sexual Medicine and Andrology Unit, Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Pasquale Gallina
- Division of Pharmacology and Toxicology, Department of Neuroscience, Psychology, Neurosurgery School of Tuscany, Drug Research and Child Health (NEUROFARBA), University of Florence, Florence, Italy
| | - Mario Maggi
- Endocrinology Unit, Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Gabriella B Vannelli
- Section of Human Anatomy and Histology, Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Annamaria Morelli
- Section of Human Anatomy and Histology, Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| |
Collapse
|
41
|
Bai N, Zhang Q, Zhang W, Liu B, Yang F, Brann D, Wang R. G-protein-coupled estrogen receptor activation upregulates interleukin-1 receptor antagonist in the hippocampus after global cerebral ischemia: implications for neuronal self-defense. J Neuroinflammation 2020; 17:45. [PMID: 32007102 PMCID: PMC6995076 DOI: 10.1186/s12974-020-1715-x] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 01/13/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND G-protein-coupled estrogen receptor (GPER/GPR30) is a novel membrane-associated estrogen receptor that can induce rapid kinase signaling in various cells. Activation of GPER can prevent hippocampal neuronal cell death following transient global cerebral ischemia (GCI), although the mechanisms remain unclear. In the current study, we sought to address whether GPER activation exerts potent anti-inflammatory effects in the rat hippocampus after GCI as a potential mechanism to limit neuronal cell death. METHODS GCI was induced by four-vessel occlusion in ovariectomized female SD rats. Specific agonist G1 or antagonist G36 of GPER was administrated using minipump, and antisense oligonucleotide (AS) of interleukin-1β receptor antagonist (IL1RA) was administrated using brain infusion kit. Protein expression of IL1RA, NF-κB-P65, phosphorylation of CREB (p-CREB), Bcl2, cleaved caspase 3, and microglial markers Iba1, CD11b, as well as inflammasome components NLRP3, ASC, cleaved caspase 1, and Cle-IL1β in the hippocampal CA1 region were investigated by immunofluorescent staining and Western blot analysis. The Duolink II in situ proximity ligation assay (PLA) was performed to detect the interaction between NLRP3 and ASC. Immunofluorescent staining for NeuN and TUNEL analysis were used to analyze neuronal survival and apoptosis, respectively. We performed Barnes maze and Novel object tests to compare the cognitive function of the rats. RESULTS The results showed that G1 attenuated GCI-induced elevation of Iba1 and CD11b in the hippocampal CA1 region at 14 days of reperfusion, and this effect was blocked by G36. G1 treatment also markedly decreased expression of the NLRP3-ASC-caspase 1 inflammasome and IL1β activation, as well as downstream NF-κB signaling, the effects reversed by G36 administration. Intriguingly, G1 caused a robust elevation in neurons of a well-known endogenous anti-inflammatory factor IL1RA, which was reversed by G36 treatment. G1 also enhanced p-CREB level in the hippocampus, a transcription factor known to enhance expression of IL1RA. Finally, in vivo IL1RA-AS abolished the anti-inflammatory, neuroprotective, and anti-apoptotic effects of G1 after GCI and reversed the cognitive-enhancing effects of G1 at 14 days after GCI. CONCLUSIONS Taken together, the current results suggest that GPER preserves cognitive function following GCI in part by exerting anti-inflammatory effects and enhancing the defense mechanism of neurons by upregulating IL1RA.
Collapse
Affiliation(s)
- Ning Bai
- Neurobiology Institute of Medical Research Center, North China University of Science and Technology, Tangshan, 063210 Hebei China
| | - Quanguang Zhang
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912 USA
| | - Wenli Zhang
- Neurobiology Institute of Medical Research Center, North China University of Science and Technology, Tangshan, 063210 Hebei China
| | - Bin Liu
- First Department of Neurology, Hospital Affiliated to North China University of Science and Technology, Tangshan, 063000 Hebei China
| | - Fang Yang
- Neurobiology Institute of Medical Research Center, North China University of Science and Technology, Tangshan, 063210 Hebei China
| | - Darrell Brann
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912 USA
| | - Ruimin Wang
- Neurobiology Institute of Medical Research Center, North China University of Science and Technology, Tangshan, 063210 Hebei China
- Key Laboratory of Dementia and Cognitive Disorder in Tangshan, North China University of Science and Technology, International Science & Technology Cooperation Base of Geriatric Medicine of China, 21 Bohai Road, Caofeidian Xincheng, Tangshan, 063210 Hebei China
| |
Collapse
|
42
|
Kumar A, Foster TC. G Protein-Coupled Estrogen Receptor: Rapid Effects on Hippocampal-Dependent Spatial Memory and Synaptic Plasticity. Front Endocrinol (Lausanne) 2020; 11:385. [PMID: 32587576 PMCID: PMC7298106 DOI: 10.3389/fendo.2020.00385] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 05/14/2020] [Indexed: 12/20/2022] Open
Abstract
In the hippocampus, estrogen regulates gene transcription linked to neuronal growth, neuroprotection, and the maintenance of memory function (1-3). The mechanism is likely to involve genomic regulation through classic estrogen receptor (ER) signaling cascades that influence transcription. Estrogens binding to classic nuclear ERs, alpha (ERα) and beta (ERβ), and have pleotropic effects on development, behavior, and neurophysiological functions, including synaptic plasticity and memory consolidation (4-7). In addition to ERα and ERβ, estrogen can also initiate activation of classical second messenger signaling cascades to influence the activity of G-proteins and a host of kinases, resulting in rapid changes in physiology (8-14). These rapid effects of estrogen are commonly mediated by membrane receptors. In the late 90s, multiple laboratories cloned cDNA/gene for an orphan G-protein-coupled receptor with very low homology with other G-protein-coupled receptors and named it G-protein-coupled receptor 30 (GPR30) (15-20). Later in 2007, the International Union of Basic and Clinical Pharmacology designated GPR30 as G protein-coupled estrogen receptor (GPER) (21); GPER is a seven-transmembrane G-protein-coupled receptor, predominantly expressed on the cell membrane (22). Interestingly, GPER is reported to mediate many of the rapid responses of estradiol in the adult brain, and is widely distributed in the mammalian brain including the plasma membrane of hippocampal neurons (23-31). GPER modulates second messenger signaling cascades involving Gαs- and Gαi/o-associated increase in cyclic adenosine monophosphate and phosphoinositide 3-kinase or Src protein kinase respectively (32, 33). Activation of GPER is also associated with phospholipase C, and the inositol receptor and ryanodine receptor-mediated increase in intracellular calcium (24, 34). This commentary is concentrated specifically on the possible rapid effects of GPER in hippocampal-dependent spatial memory function and synaptic plasticity.
Collapse
|
43
|
Evans PD. Rapid signalling responses via the G protein-coupled estrogen receptor, GPER, in a hippocampal cell line. Steroids 2019; 152:108487. [PMID: 31499073 DOI: 10.1016/j.steroids.2019.108487] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 08/19/2019] [Accepted: 09/03/2019] [Indexed: 01/14/2023]
Abstract
The rapid non-genomic actions of 17β-estradiol in multiple tissues, including the nervous system, may involve the activation of the G-protein-coupled receptor, GPER. Different signalling pathways have been suggested to be activated by GPER in different cell lines and tissues. Controversially, GPER has also been suggested to be activated by the mineralocorticoid aldosterone, and by the non-steroidal diphenylacrylamide compound, STX, in some preparations. Evidence for the ability of the GPER agonist, G-1, and for aldosterone in the presence of the mineralocorticoid receptor antagonist, eplerenone, to potentiate forskolin-stimulated cyclic AMP levels in the hippocampal clonal cell line, mHippoE-18 is reviewed. The effects of both agents are blocked by the GPER antagonist G36, by PTX, (suggesting the involvement of Gi/o G proteins), by BAPTA-AM, (suggesting they are calcium sensitive), by wortmannin (suggesting an involvement of PI3Kinase) and by soluble amyloid-β peptides. STX also stimulates cyclic AMP levels in mHippoE-18 cells and these effects are blocked by G36 and PTX, as well as by amyloid-β peptides. This suggests that both aldosterone and STX may be capable of activating GPER in mHippoE-18 cells. Possible molecular mechanisms that may underlie these effects are discussed, together with possible forward directions for research on rapid non-genomic signalling by GPER, emphasising the importance of understanding the spatio-temporal aspects of its signalling in various tissues.
Collapse
Affiliation(s)
- Peter D Evans
- The Signalling Laboratory, The Babraham Institute, The Babraham Research Campus, Cambridge CB22 3AT, UK.
| |
Collapse
|
44
|
Bannister E. There is increasing evidence to suggest that brain inflammation could play a key role in the aetiology of psychiatric illness. Could inflammation be a cause of the premenstrual syndromes PMS and PMDD? Post Reprod Health 2019; 25:157-161. [PMID: 31630609 DOI: 10.1177/2053369119875386] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
45
|
Ariyani W, Miyazaki W, Koibuchi N. A Novel Mechanism of S-equol Action in Neurons and Astrocytes: The Possible Involvement of GPR30/GPER1. Int J Mol Sci 2019; 20:ijms20205178. [PMID: 31635400 PMCID: PMC6829462 DOI: 10.3390/ijms20205178] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 10/15/2019] [Accepted: 10/17/2019] [Indexed: 12/16/2022] Open
Abstract
S-equol is a major bacterial metabolite of the soy isoflavone daidzein. It is known to be a phytoestrogen that acts by binding to the nuclear estrogen receptors (ERs) that are expressed in various brain regions, including the cerebellum. However, the effects of S-equol on cerebellar development and function have not yet been extensively studied. In this study, the effects of S-equol were evaluated using a mouse primary cerebellar culture, Neuro-2A clonal cells, and an astrocyte-enriched culture. S-equol augmented the dendrite arborization of Purkinje cells induced by triiodothyronine (T3) and the neurite growth of Neuro-2A cell differentiation. Such augmentation was suppressed by G15, a selective G-protein coupled ER (GPR30) antagonist, and ICI 182,780, an antagonist for ERs in both cultures. On the other hand, in astrocytes, S-equol induced cell proliferation and cell migration with an increase in the phosphorylated extracellular-signal-regulated kinase 1/2 and F-actin rearrangements. Such effects were suppressed by G15, but not by ICI. These findings indicated that S-equol may enhanced cerebellar development by affecting both neurons and astrocytes through several signaling pathways, including GPR30 and ERs. We here report a novel mechanism of S-equol in cerebellar development that may provide a novel possibility to use S-equol supplementation during development.
Collapse
Affiliation(s)
- Winda Ariyani
- Department of Integrative Physiology, Graduate School of Medicine, Gunma University, 3-39-22 Showa-machi Maebashi, Gunma 371-8511, Japan.
| | - Wataru Miyazaki
- Department of Integrative Physiology, Graduate School of Medicine, Gunma University, 3-39-22 Showa-machi Maebashi, Gunma 371-8511, Japan.
- Department of Bioscience and Laboratory Medicine, Graduate School of Health Science, Hirosaki University, 66-1 Hon-cho Aomori, Hirosaki 036-8564, Japan.
| | - Noriyuki Koibuchi
- Department of Integrative Physiology, Graduate School of Medicine, Gunma University, 3-39-22 Showa-machi Maebashi, Gunma 371-8511, Japan.
| |
Collapse
|
46
|
Evans PD. Aldosterone, STX and amyloid-β 1-42 peptides modulate GPER (GPR30) signalling in an embryonic mouse hippocampal cell line (mHippoE-18). Mol Cell Endocrinol 2019; 496:110537. [PMID: 31404576 DOI: 10.1016/j.mce.2019.110537] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 08/05/2019] [Accepted: 08/08/2019] [Indexed: 01/02/2023]
Abstract
The GPCR, GPER, mediates many of the rapid, non-genomic actions of 17β-estradiol in multiple tissues, including the nervous system. Controversially, it has also been suggested to be activated by aldosterone, and by the non-steroidal diphenylacrylamide compound, STX, in some preparations. Here, the ability of the GPER agonist, G-1, and aldosterone in the presence of the mineralocorticoid receptor antagonist, eplerenone, to potentiate forskolin-stimulated cyclic AMP levels in the hippocampal clonal cell line, mHippoE-18, are compared. Both stimulatory effects are blocked by the GPER antagonist G36, by PTX, (suggesting the involvement of Gi/o G proteins), by BAPTA-AM, (suggesting they are calcium sensitive), by wortmannin (suggesting an involvement of PI3Kinase) and by soluble amyloid-β peptides. STX also stimulates cyclic AMP levels in mHippoE-18 cells and these effects are blocked by G36 and PTX, as well as by amyloid-β peptides. This suggests that both aldosterone and STX may modulate GPER signalling in mHippoE-18 cells.
Collapse
Affiliation(s)
- Peter D Evans
- The Signalling Laboratory, The Babraham Institute, The Babraham Research Campus, Cambridge, CB22 3AT, UK.
| |
Collapse
|
47
|
Roque C, Mendes-Oliveira J, Duarte-Chendo C, Baltazar G. The role of G protein-coupled estrogen receptor 1 on neurological disorders. Front Neuroendocrinol 2019; 55:100786. [PMID: 31513775 DOI: 10.1016/j.yfrne.2019.100786] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 09/02/2019] [Accepted: 09/07/2019] [Indexed: 02/06/2023]
Abstract
G protein-coupled estrogen receptor 1 (GPER) is a membrane-associated estrogen receptor (ER) associated with rapid estrogen-mediated effects. Over recent years GPER emerged has a potential therapeutic target to induce neuroprotection, avoiding the side effects elicited by the activation of classical ERs. The putative neuroprotection triggered by GPER selective activation was demonstrated in mood disorders, Alzheimer's disease or Parkinson's disease of male and female in vivo rodent models. In others, like ischemic stroke, the results are contradictory and currently there is no consensus on the role played by this receptor. However, it seems clear that sex is a biological variable that may impact the results. The major objective of this review is to provide an overview about the physiological effects of GPER in the brain and its putative contribution in neurodegenerative disorders, discussing the data about the signaling pathways involved, as well as, the diverse effects observed.
Collapse
Affiliation(s)
- C Roque
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - J Mendes-Oliveira
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - C Duarte-Chendo
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - G Baltazar
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal; Faculty of Health Sciences, University of Beira Interior, Covilhã, Portugal.
| |
Collapse
|
48
|
G protein-coupled estrogen receptor 1 negatively regulates the proliferation of mouse-derived neural stem/progenitor cells via extracellular signal-regulated kinase pathway. Brain Res 2019; 1714:158-165. [PMID: 30797747 DOI: 10.1016/j.brainres.2019.02.024] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 02/14/2019] [Accepted: 02/19/2019] [Indexed: 12/20/2022]
Abstract
G protein-coupled estrogen receptor 1 (GPER1, also known as GPR30) has been reported to play a wide range of function in the central nervous system (CNS). However, whether GPER1 is expressed by neural stem/progenitor cells (NSPCs) and its role has not been established. Here, we found the expression of GPER1 in mouse-derived NSPCs via western blot and immunofluorescent staining. Moreover, we revealed that specific activation of GPER1 by the agonist G1 decreased the proliferation of NSPCs in a dose-dependent manner. The neurosphere formation assay and Ki67 staining further demonstrated that activation of GPER1 inhibited the proliferation of NSPCs. Additionally, the inhibitory effect of G1 on the proliferation of NSPCs could be blocked by the specific GPER1 antagonist G15. Intriguingly, ERK pathway was involved in the negative effect of GPER1 on the proliferation of NSPCs, because the phosphorylation level of ERK in NSPCs was remarkably decreased during G1 treatment. However, the antagonist G15 reversed the down-regulated level of p-ERK. Knock-down GPER1 also reversed the inhibitory effect of G1 on NSPCs proliferation. Together, our results provide the first evidence that GPER1 is expressed by NSPCs and its activation negatively modulates the proliferation of NSPCs, highlighting the importance of GPER1 in regulating NSPC behaviors.
Collapse
|
49
|
Meitzen J, Britson KA, Tuomela K, Mermelstein PG. The expression of select genes necessary for membrane-associated estrogen receptor signaling differ by sex in adult rat hippocampus. Steroids 2019; 142:21-27. [PMID: 28962849 PMCID: PMC5874170 DOI: 10.1016/j.steroids.2017.09.012] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 09/14/2017] [Accepted: 09/22/2017] [Indexed: 12/21/2022]
Abstract
17β-estradiol can rapidly modulate neuron function via membrane estrogen receptors (ERs) in a sex-specific manner. For example, female rat hippocampal neurons express palmitoylated versions of ERα and ERβ that associate with the plasma membrane. These membrane-associated ERs are organized by caveolin proteins into functional signaling microdomains with metabotropic glutamate receptors (mGluRs). ER/mGluR signaling mediates several sex-specific estradiol actions on hippocampal neuron function. An important unanswered question regards the mechanism by which sex-specific membrane-associated ER signaling is generated, especially since it has been previously demonstrated that mGluR action is not sex-specific. One possibility is that the genes necessary for the ER membrane complex are differentially expressed between males and females, including genes that encode ERα and β, caveolin 1 and 3, and/or the palmitoylacyltransferases DHHC-7 and -21. Thus we used qPCR to test the hypothesis that these genes show sex differences in expression in neonatal and adult rat hippocampus. As an additional control we tested the expression of the 20 other DHHC palmitoylacyltransferases with no known connections to ER. In neonatal hippocampus, no sex differences were detected in gene expression. In adult hippocampus, the genes that encode caveolin 1 and DHHC-7 showed decreased expression in females compared to males. Thus, select genes differ by sex at specific developmental stages, arguing for a more nuanced model than simple widespread perinatal emergence of sex differences in all genes enabling sex-specific estradiol action. These findings enable the generation of new hypotheses regarding the mechanisms by which sex differences in membrane-associated ER signaling are programmed.
Collapse
Affiliation(s)
- John Meitzen
- Dept. of Biological Sciences, North Carolina State University, Raleigh, NC, United States; W.M. Keck Center for Behavioral Biology, North Carolina State University, Raleigh, NC, United States; Center for Human Health and the Environment, North Carolina State University, Raleigh, NC, United States; Comparative Medicine Institute, North Carolina State University, Raleigh, NC, United States.
| | - Kyla A Britson
- Cellular and Molecular Medicine Graduate Program, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Krista Tuomela
- Medical College of Wisconsin, Milwaukee, WI, United States
| | - Paul G Mermelstein
- Dept. of Neuroscience, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
50
|
Choudhury N, Sikdar SK. 17β-estradiol potentiates TREK1 channel activity through G protein-coupled estrogen receptor. J Steroid Biochem Mol Biol 2018; 183:94-105. [PMID: 29883692 DOI: 10.1016/j.jsbmb.2018.06.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Revised: 05/10/2018] [Accepted: 06/04/2018] [Indexed: 11/21/2022]
Abstract
TWIK-related potassium channel 1 (TREK1), a two-pore domain potassium channel, is modulated by various hormones and neurotransmitters by activation of membrane receptor - coupled second messengers. 17β-estradiol is a neuromodulator capable of regulating several cellular processes including the activity of ion channels, in a rapid and non-genomic manner. The G protein-coupled estrogen receptor (GPER) is known to facilitate rapid actions of 17β-estradiol, though its role in modulation of ion channels is not widely explored. Several studies have shown both TREK1 and 17β-estradiol to be neuromodulatory but the interaction between them is not known. In the present study, using single channel cell-attached patch clamp electrophysiology in HEK293 cells, we show that 17β-estradiol increases the activity of hTREK1 channel by acting through hGPER and increasing the channel opening probability within minutes. The potentiation induced by 17β-estradiol is pertussis toxin - sensitive involving action of Gβγ subunits while the inhibitory effect of cAMP-PKA pathway on TREK1 is reduced. Protein phosphatases were also found to be important for the action of 17β-estradiol, which in concert with reduced activity of PKA, may alter the phosphorylation state of the channel and thus increase channel activity. Mutational studies revealed the serines at positions 315 and 348 in the C-terminal domain of hTREK1 to be the target sites for dephosphorylation induced by 17β-estradiol action through hGPER. Elucidation of the pathway for the potentiating action of 17β-estradiol via hGPER on hTREK1 channel activity will help us understand better one of the many possible neuroprotective mechanisms of 17β-estradiol and hTREK1 channel.
Collapse
Affiliation(s)
- Nasreen Choudhury
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore, 560012, Karnataka, India.
| | - Sujit Kumar Sikdar
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore, 560012, Karnataka, India.
| |
Collapse
|