1
|
Xue Y, Zhang Y, Wu Y, Zhao T. Activation of GPER-1 Attenuates Traumatic Brain Injury-Induced Neurological Impairments in Mice. Mol Neurobiol 2024; 61:5614-5627. [PMID: 38217667 DOI: 10.1007/s12035-024-03919-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Accepted: 01/01/2024] [Indexed: 01/15/2024]
Abstract
This study aimed to investigate the effects of G1-activated G protein-coupled estrogen receptor 1 (GPER1) on neurological impairments and neuroinflammation in traumatic brain injury (TBI) mice. The controlled cortical impingement (CCI) method was used to establish the TBI model. The mice were subjected to ovariectomy (OVX) for two weeks prior to modeling. GPER1 agonist G1 was administered by intracerebroventricular injection. Brain tissue water content was detected by wet/dry method, and blood-brain barrier damage was detected by Evans blue extravasation. The neurological impairments in mice were evaluated by open field test, Y-maze test, nest-building test, object location memory test and novel object recognition test. Ionized calcium-binding adapter molecule 1 (Iba1) staining was used to indicate the activation of microglia. Expression of M1/M2-type microglia markers and inflammatory factors were evaluated by ELISA and qRT-PCR. The G1 administration significantly reduced cerebral edema and Evans blue extravasation at injury ipsilateral cortex and basal ganglia in TBI mice. Activation of GPER1 by G1 improved the anxiety behavior and the cognitive dysfunction of mice induced by TBI. G1 administration significantly decreased Iba1-positive staining cells and the mRNA levels of CD86, macrophage cationic peptide 1 (Mcp-1), nitric oxide synthase 2 (Nos2), interleukin 1 beta (IL-1β), and macrophage inflammatory protein-2 (MIP-2), while increased the mRNA levels of interleukin 10 (IL-10), arginase1 (Arg-1) and CD206. Activation of GPER1 through G1 administration has the potential to ameliorate cognitive dysfunction induced by TBI in mice. It may also inhibit the activation of M1 microglia in cortical tissue resulting from TBI, while promoting the activation of M2 microglia and contributing to the regulation of inflammatory responses.
Collapse
Affiliation(s)
- Yafei Xue
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, No. 569 Xinsi Road, Baqiao District, Xi'an, 710038, Shaanxi, China
| | - Yunze Zhang
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, No. 569 Xinsi Road, Baqiao District, Xi'an, 710038, Shaanxi, China
| | - Yingxi Wu
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, No. 569 Xinsi Road, Baqiao District, Xi'an, 710038, Shaanxi, China.
| | - Tianzhi Zhao
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, No. 569 Xinsi Road, Baqiao District, Xi'an, 710038, Shaanxi, China.
| |
Collapse
|
2
|
Dong H, Zeng X, Xu J, He C, Sun Z, Liu L, Huang Y, Sun Z, Cao Y, Peng Z, Qiu YA, Yu T. Advances in immune regulation of the G protein-coupled estrogen receptor. Int Immunopharmacol 2024; 136:112369. [PMID: 38824903 DOI: 10.1016/j.intimp.2024.112369] [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: 02/16/2024] [Revised: 04/12/2024] [Accepted: 05/27/2024] [Indexed: 06/04/2024]
Abstract
Estrogen and related receptors have been shown to have a significant impact on human development, reproduction, metabolism and immune regulation and to play a critical role in tumor development and treatment. Traditionally, the nuclear estrogen receptors (nERs) ERα and ERβ have been thought to be involved in mediating the estrogenic effects. However, our group and others have previously demonstrated that the G protein-coupled estrogen receptor (GPER) is the third independent ER, and estrogen signaling mediated by GPER is known to play an important role in normal physiology and a variety of abnormal diseases. Interestingly, recent studies have progressively revealed GPER involvement in the maintenance of the normal immune system, abnormal immune diseases, and inflammatory lesions, which may be of significant clinical value primarily in the immunotherapy of tumors. In this article, we review current advances in GPER-related immunomodulators and provide a theoretical basis and potential clinical targets to ameliorate immune-related diseases and immunotherapy for tumors.
Collapse
Affiliation(s)
- Hanzhi Dong
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330000, China
| | - Xiaoqiang Zeng
- Jiangxi Medical College, Nanchang University, Nanchang 330000, China
| | - Jiawei Xu
- Department of Breast Surgery, Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Jiangxi Clinical Research Center for Cancer, Nanchang 330029, China
| | - Chongwu He
- Department of Breast Surgery, Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Jiangxi Clinical Research Center for Cancer, Nanchang 330029, China
| | - Zhengkui Sun
- Department of Breast Surgery, Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Jiangxi Clinical Research Center for Cancer, Nanchang 330029, China
| | - Liyan Liu
- Department of Pharmacy, Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Jiangxi Clinical Research Center for Cancer, Nanchang 330029, China
| | - Yanxiao Huang
- Jiangxi Medical College, Nanchang University, Nanchang 330000, China
| | - Zhe Sun
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330000, China
| | - Yuan Cao
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330000, China
| | - Zhiqiang Peng
- Department of Lymphohematology, Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Jiangxi Clinical Research Center for Cancer, Nanchang 330029, China.
| | - Yu-An Qiu
- Department of Critical Care Medicine, Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Jiangxi Clinical Research Center for Cancer, Nanchang 330029, China.
| | - Tenghua Yu
- Department of Breast Surgery, Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Jiangxi Clinical Research Center for Cancer, Nanchang 330029, China.
| |
Collapse
|
3
|
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
|
4
|
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
|
5
|
Amirkhosravi L, Khaksari M, Sanjari M, Khorasani P. The nongenomic neuroprotective effects of estrogen, E2-BSA, and G1 following traumatic brain injury: PI3K/Akt and histopathological study. Horm Mol Biol Clin Investig 2024; 45:1-15. [PMID: 38507353 DOI: 10.1515/hmbci-2023-0066] [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: 09/19/2023] [Accepted: 02/28/2024] [Indexed: 03/22/2024]
Abstract
OBJECTIVES Studies suggest that both genomic and nongenomic pathways are involved in mediating the salutary effects of steroids following traumatic brain injury (TBI). This study investigated the nongenomic effects of 17β-estradiol (E2) mediated by the PI3K/p-Akt pathway after TBI. METHODS Ovariectomized rats were apportioned to E2, 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 vehicles were injected before the induction of TBI and injection of drugs. Diffuse TBI was induced by the Marmarou model. Evans blue (EBC, 5 h), brain water contents (BWC), histopathological changes, and brain PI3K and p-Akt protein expressions were measured 24 h after TBI. The veterinary comma scale (VCS) was assessed before and at different times after TBI. RESULTS The results showed a reduction in BWC and EBC and increased VCS in the E2, E2-BSA, and G1 groups. Also, E2, E2-BSA, and G1 reduced brain edema, inflammation, and apoptosis. The ICI and G15 inhibited the beneficial effects of E2, E2-BSA, and G1 on these parameters. All drugs, following TBI, prevented the reduction of brain PI3K/p-Akt expression. The individual or combined use of ICI and G15 eliminated the beneficial effects of E2, E2-BSA, and G1 on PI3K/p-Akt expressions. CONCLUSIONS These findings indicated that PI3K/p-Akt pathway plays a critical role in mediating the salutary effects of estradiol on histopathological changes and neurological outcomes following TBI, suggesting that GPER and classic ERs are involved in regulating the expression of PI3K/p-Akt.
Collapse
Affiliation(s)
- Ladan Amirkhosravi
- Endocrinology and Metabolism Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Khaksari
- Physiology Research Center, Institute of Neuropharmacology, 48463 Kerman University of Medical Sciences , Kerman, Iran
| | - Mojgan Sanjari
- Endocrinology and Metabolism Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Parisa Khorasani
- Pathology and Stem Cell Research Center, 48463 Kerman University of Medical Sciences , Kerman, Iran
| |
Collapse
|
6
|
Jouffre B, Acramel A, Jacquot Y, Daulhac L, Mallet C. GPER involvement in inflammatory pain. Steroids 2023; 200:109311. [PMID: 37734514 DOI: 10.1016/j.steroids.2023.109311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 09/11/2023] [Accepted: 09/18/2023] [Indexed: 09/23/2023]
Abstract
Chronic pain is a worldwide refractory health disease that causes major financial and emotional burdens and that is devastating for individuals and society. One primary source of pain is inflammation. Current treatments for inflammatory pain are weakly effective, although they usually replace analgesics, such as opioids and non-steroidal anti-inflammatory drugs, which display serious side effects. Emerging evidence indicates that the membrane G protein-coupled estrogen receptor (GPER) may play an important role in the regulation of inflammation and pain. Herein, we focus on the consequences of pharmacological and genetic GPER modulation in different animal models of inflammatory pain. We also provide a brief overview of the putative mechanisms including the direct action of GPER on pain transmission and inflammation.
Collapse
Affiliation(s)
- Baptiste Jouffre
- Université Clermont Auvergne, Inserm U1107 Neuro-Dol, Pharmacologie Fondamentale et Clinique de la Douleur, Clermont-Ferrand, France; ANALGESIA Institute, Faculty of Medicine, 63000 Clermont-Ferrand, France
| | - Alexandre Acramel
- CiTCoM, CNRS - UMR 8038, INSERM U1268, Faculty of Pharmacy of Paris, University Paris Cité, 75270 Paris Cedex 06, France; Department of Pharmacy, Institut Curie, 75248 Paris Cedex 06, France
| | - Yves Jacquot
- CiTCoM, CNRS - UMR 8038, INSERM U1268, Faculty of Pharmacy of Paris, University Paris Cité, 75270 Paris Cedex 06, France
| | - Laurence Daulhac
- Université Clermont Auvergne, Inserm U1107 Neuro-Dol, Pharmacologie Fondamentale et Clinique de la Douleur, Clermont-Ferrand, France; ANALGESIA Institute, Faculty of Medicine, 63000 Clermont-Ferrand, France
| | - Christophe Mallet
- Université Clermont Auvergne, Inserm U1107 Neuro-Dol, Pharmacologie Fondamentale et Clinique de la Douleur, Clermont-Ferrand, France; ANALGESIA Institute, Faculty of Medicine, 63000 Clermont-Ferrand, France.
| |
Collapse
|
7
|
Ferreira LO, de Souza RD, Teixeira LL, Pinto LC, Rodrigues JCM, Martins-Filho AJ, da Costa ET, Hamoy M, Lopes DCF. The GPER1 agonist G1 reduces brain injury and improves the qEEG and behavioral outcome of experimental ischemic stroke. J Neuropathol Exp Neurol 2023; 82:787-797. [PMID: 37558387 DOI: 10.1093/jnen/nlad061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/11/2023] Open
Abstract
Stroke is one of the principal cerebrovascular diseases in human populations and contributes to a majority of the functional impairments in the elderly. Recent discoveries have led to the inclusion of electroencephalography (EEG) in the complementary prognostic evaluation of patients. The present study describes the EEG, behavioral, and histological changes that occur following cerebral ischemia associated with treatment by G1, a potent and selective G protein-coupled estrogen receptor 1 (GPER1) agonist in a rat model. Treatment with G1 attenuated the neurological deficits induced by ischemic stroke from the second day onward, and reduced areas of infarction. Treatment with G1 also improved the total brainwave power, as well as the theta and alpha wave activity, specifically, and restored the delta band power to levels similar to those observed in the controls. Treatment with G1 also attenuated the peaks of harmful activity observed in the EEG indices. These improvements in brainwave activity indicate that GPER1 plays a fundamental role in the mediation of cerebral injury and in the behavioral outcome of ischemic brain injuries, which points to treatment with G1 as a potential pharmacological strategy for the therapy of stroke.
Collapse
Affiliation(s)
- Luan Oliveira Ferreira
- Laboratory of Experimental Neuropathology, Joao de Barros Barreto University Hospital, Federal University of Pará, Belém, Brazil
| | - Rafael Dias de Souza
- Laboratory of Experimental Neuropathology, Joao de Barros Barreto University Hospital, Federal University of Pará, Belém, Brazil
| | - Leonan Lima Teixeira
- Laboratory of Experimental Neuropathology, Joao de Barros Barreto University Hospital, Federal University of Pará, Belém, Brazil
| | - Laine Celestino Pinto
- Laboratory of Experimental Neuropathology, Joao de Barros Barreto University Hospital, Federal University of Pará, Belém, Brazil
| | - Joao Cleiton Martins Rodrigues
- Laboratory of Experimental Neuropathology, Joao de Barros Barreto University Hospital, Federal University of Pará, Belém, Brazil
| | | | - Edmar Tavares da Costa
- Laboratory of Experimental Neuropathology, Joao de Barros Barreto University Hospital, Federal University of Pará, Belém, Brazil
| | - Moisés Hamoy
- Laboratory of Pharmacology and Toxicology of Natural Products, Biological Sciences Institute, Federal University of Pará, Belém, Brazil
| | - Dielly Catrina Favacho Lopes
- Laboratory of Experimental Neuropathology, Joao de Barros Barreto University Hospital, Federal University of Pará, Belém, Brazil
| |
Collapse
|
8
|
Zhong X, Sun Y, Lu Y, Xu L. Immunomodulatory role of estrogen in ischemic stroke: neuroinflammation and effect of sex. Front Immunol 2023; 14:1164258. [PMID: 37180115 PMCID: PMC10167039 DOI: 10.3389/fimmu.2023.1164258] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Accepted: 04/12/2023] [Indexed: 05/15/2023] Open
Abstract
Although estrogen is predominantly related to the maintenance of reproductive functioning in females, it mediates various physiological effects in nearly all tissues, especially the central nervous system. Clinical trials have revealed that estrogen, especially 17β-estradiol, can attenuate cerebral damage caused by an ischemic stroke. One mechanism underlying this effect of 17β-estradiol is by modulating the responses of immune cells, indicating its utility as a novel therapeutic strategy for ischemic stroke. The present review summarizes the effect of sex on ischemic stroke progression, the role of estrogen as an immunomodulator in immune reactions, and the potential clinical value of estrogen replacement therapy. The data presented here will help better understand the immunomodulatory function of estrogen and may provide a basis for its novel therapeutic use in ischemic stroke.
Collapse
Affiliation(s)
- Xiaojun Zhong
- Department of Neurosurgery, Zhejiang Rongjun Hospital, Jiaxing, China
| | - Yulin Sun
- Department of Neurosurgery, Zhejiang Rongjun Hospital, Jiaxing, China
| | - Yajun Lu
- Department of Internal Medicine, Sunto Women & Children’s Hospital, Jiaxing, China
| | - Lei Xu
- Department of Neurology, Zhejiang Rongjun Hospital, Jiaxing, China
| |
Collapse
|
9
|
Non-genomic Effect of Estradiol on the Neurovascular Unit and Possible Involvement in the Cerebral Vascular Accident. Mol Neurobiol 2023; 60:1964-1985. [PMID: 36596967 DOI: 10.1007/s12035-022-03178-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 12/16/2022] [Indexed: 01/05/2023]
Abstract
Cerebrovascular diseases, such as ischemic cerebral vascular accident (CVA), are responsible for causing high rates of morbidity, mortality, and disability in the population. The neurovascular unit (NVU) during and after ischemic CVA plays crucial roles in cell regulation and preservation, the immune and inflammatory response, and cell and/or tissue survival and repair. Cellular responses to 17β-estradiol (E2) can be triggered by two mechanisms: one called classical or genomic, which is due to the activation of the "classical" nuclear estrogen receptors α (ERα) and β (ERβ), and the non-genomic or rapid mechanism, which is due to the activation of the G protein-coupled estrogen receptor 1 (GPER) that is located in the plasma membrane and some in intracellular membranes, such as in the Golgi apparatus and endoplasmic reticulum. Nuclear receptors can regulate gene expression and cellular functions. On the contrary, activating the GPER by E2 and/or its G-1 agonist triggers several rapid cell signaling pathways. Therefore, E2 or its G-1 agonist, by mediating GPER activation and/or expression, can influence several NVU cell types. Most studies argue that the activation of the GPER may be used as a potential therapeutic target in various pathologies, such as CVA. Thus, with this review, we aimed to summarize the existing literature on the role of GPER mediated by E2 and/or its agonist G-1 in the physiology and pathophysiology of NVU.
Collapse
|
10
|
da Silva FER, Cordeiro RC, de Carvalho Lima CN, Cardozo PL, Vasconcelos GS, Monte AS, Sanders LLO, Vasconcelos SMM, de Lucena DF, Cruz BF, Nicolato R, Seeman MV, Ribeiro FM, Macedo DS. Sex and the Estrous-Cycle Phase Influence the Expression of G Protein-Coupled Estrogen Receptor 1 (GPER) in Schizophrenia: Translational Evidence for a New Target. Mol Neurobiol 2023; 60:3650-3663. [PMID: 36917419 DOI: 10.1007/s12035-023-03295-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 02/24/2023] [Indexed: 03/16/2023]
Abstract
Schizophrenia is a mental disorder with sex bias in disease onset and symptom severity. Recently, it was observed that females present more severe symptoms in the perimenstrual phase of the menstrual cycle. The administration of estrogen also alleviates schizophrenia symptoms. Despite this, little is known about symptom fluctuation over the menstrual cycle and the underlying mechanisms. To address this issue, we worked with the two-hit schizophrenia animal model induced by neonatal exposure to a virus-like particle, Poly I:C, associated with peripubertal unpredictable stress exposure. Prepulse inhibition of the startle reflex (PPI) in male and female mice was considered analogous to human schizophrenia-like behavior. Female mice were studied in the proestrus (high-estrogen estrous cycle phase) and diestrus (low-estrogen phase). Additionally, we evaluated the hippocampal mRNA expression of estrogen synthesis proteins; TSPO and aromatase; and estrogen receptors ERα, ERβ, and GPER. We also collected peripheral blood mononuclear cells (PBMCs) from male and female patients with schizophrenia and converted them to induced microglia-like cells (iMGs) to evaluate the expression of GPER. We observed raised hippocampal expression of GPER in two-hit female mice at the proestrus phase without PPI deficits and higher levels of proteins related to estrogen synthesis, TSPO, and aromatase. In contrast, two-hit adult males with PPI deficits presented lower hippocampal mRNA expression of TSPO, aromatase, and GPER. iMGs from male and female patients with schizophrenia showed lower mRNA expression of GPER than controls. Therefore, our results suggest that GPER alterations constitute an underlying mechanism for sex influence in schizophrenia.
Collapse
Affiliation(s)
- Francisco Eliclécio Rodrigues da Silva
- Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceara, Rua Cel. Nunes de Melo 1000, 60430-275, CE, Fortaleza, Brazil
| | - Rafaela Carneiro Cordeiro
- Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceara, Rua Cel. Nunes de Melo 1000, 60430-275, CE, Fortaleza, Brazil.,University of Texas Health Science Center at Houston, Houston, USA
| | - Camila N de Carvalho Lima
- Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceara, Rua Cel. Nunes de Melo 1000, 60430-275, CE, Fortaleza, Brazil.,University of Texas Health Science Center at Houston, Houston, USA
| | - Pablo Leal Cardozo
- Department of Biochemistry and Immunology, Institute of Biological Sciences (ICB), Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Germana Silva Vasconcelos
- Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceara, Rua Cel. Nunes de Melo 1000, 60430-275, CE, Fortaleza, Brazil
| | - Aline Santos Monte
- Health Science Institute, University of International Integration of Afro-Brazilian Lusophony UNILAB, Redenção, Brazil
| | - Lia Lira Olivier Sanders
- Course of Medicine, Centro Universitário Christus-Unichristus, Fortaleza, Brazil.,Department of Clinical Medicine, Federal University of Ceara, Fortaleza, Brazil
| | - Silvânia Maria Mendes Vasconcelos
- Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceara, Rua Cel. Nunes de Melo 1000, 60430-275, CE, Fortaleza, Brazil
| | - David Freitas de Lucena
- Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceara, Rua Cel. Nunes de Melo 1000, 60430-275, CE, Fortaleza, Brazil
| | - Breno Fiuza Cruz
- Department of Mental Health, Faculty of Medicine, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Rodrigo Nicolato
- Department of Mental Health, Faculty of Medicine, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Mary V Seeman
- Department of Psychiatry, University of Toronto, Toronto, Canada
| | - Fabíola Mara Ribeiro
- Department of Biochemistry and Immunology, Institute of Biological Sciences (ICB), Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Danielle S Macedo
- Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceara, Rua Cel. Nunes de Melo 1000, 60430-275, CE, Fortaleza, Brazil. .,National Institute for Translational Medicine (INCT-TM, CNPq), São Paulo, Brazil.
| |
Collapse
|
11
|
Lu D, Ping H, Wei C, Fang W, Zhang Y, Wu Y, Xue Y, Wang B, Qu Y, Zhao T. Unruptured anterior Inferior cerebellar artery aneurysm following stereotactic irradiation for vestibular schwannoma: Case report and literature review. Front Surg 2023; 10:1082265. [PMID: 36843988 PMCID: PMC9949606 DOI: 10.3389/fsurg.2023.1082265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 01/16/2023] [Indexed: 02/11/2023] Open
Abstract
Background The clinical features and therapeutic measures of vestibular schwannoma (VS) radiation-related aneurysm (RRA) have not been well described. We reported the first VS RRA case admitted for acute anterior inferior cerebella artery (AICA) ischemic symptoms. Literature was reviewed to present the research fruits about VS RRAs, and some therapeutic advices were given. Materials and methods A 54-year-old woman who had undergone GKS 10 years previously for a right VS was admitted to our hospital in 2018 because of sudden onset of severe vertigo and vomiting, accompanied with unsteady gait. During tumor resection, a dissecting aneurysm arose from the main trunk of AICA was encountered accidently within the tumor. The aneurysm was successfully treated with direct clip ligation, sparing the parent vessel. Data about this case were combined with those of other 11 radiation-related AICA aneurysm cases retrieved from the current literature. The following parameters were evaluated: Age, Sex, Diagnostic method, Location of aneurysm, Age of radiotherapy (Years)/Latency, Rupture, x-ray dosage, Type of radiotherapy, History of surgical resection of VS, Aneurysm Type, Morphology, Number, Treatment, Operative complications, Sequela, Outcome. VS RRAs mainly occurred in women (75%) with a median age of 62.5 years and were mainly located on AICA. Ruptured aneurysms accounted for 75.0% of the total cases. This paper reported the first VS case admitted with acute AICA ischemic symptoms. Cases with sacciform-like, irregular and fusiform-shaped aneurysms accounted for 50.0%, 25.0% and 25.0% of the total, respectively. After surgical treatment, 75.0% patients recovered, except for 3 patients who developed new ischemic consequence. Conclusion Patients should be informed of the risk of RRAs after receiving radiotherapy for VS. In these patients, RRAs should be suspected when subarachnoid hemorrhage or AICA ischemic symptoms occurred. Active intervention should be conducted considering the high instability and bleeding rate of VS RRAs.
Collapse
Affiliation(s)
| | | | | | - Wei Fang
- Department of Neurosurgery, The Second Affiliated Hospital, Air Force Military Medical University, Xi’an, China
| | - Yunze Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, Air Force Military Medical University, Xi’an, China
| | - Yingxi Wu
- Department of Neurosurgery, The Second Affiliated Hospital, Air Force Military Medical University, Xi’an, China
| | - Yafei Xue
- Department of Neurosurgery, The Second Affiliated Hospital, Air Force Military Medical University, Xi’an, China
| | - Bao Wang
- Correspondence: Tianzhi Zhao Yan Qu Bao Wang
| | - Yan Qu
- Correspondence: Tianzhi Zhao Yan Qu Bao Wang
| | | |
Collapse
|
12
|
Han Y, Gu S, Li Y, Qian X, Wang F, Huang JH. Neuroendocrine pathogenesis of perimenopausal depression. Front Psychiatry 2023; 14:1162501. [PMID: 37065890 PMCID: PMC10098367 DOI: 10.3389/fpsyt.2023.1162501] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 03/13/2023] [Indexed: 04/18/2023] Open
Abstract
With the development of social economics and the increase of working pressure, more and more women are suffering from long-term serious stress and showing symptoms of perimenopausal depression (PMD). The incidence rate of PMD is increasing, and the physical and mental health are seriously affected. However, due to the lack of accurate knowledge of pathophysiology, its diagnosis and treatment cannot be accurately executed. By consulting the relevant literature in recent years, this paper elaborates the neuroendocrine mechanism of perimenopausal depression from the aspects of epigenetic changes, monoamine neurotransmitter and receptor hypothesis, glial cell-induced neuroinflammation, estrogen receptor, interaction between HPA axis and HPG axis, and micro-organism-brain gut axis. The purpose is to probe into new ways of treatment of PMD by providing new knowledge about the neuroendocrine mechanism and treatment of PMD.
Collapse
Affiliation(s)
- Yuping Han
- Department of Psychology, Medical School, Jiangsu University, Zhenjiang, China
| | - Simeng Gu
- Department of Psychology, Medical School, Jiangsu University, Zhenjiang, China
- *Correspondence: Simeng Gu,
| | - Yumeng Li
- Department of Psychology, Medical School, Jiangsu University, Zhenjiang, China
| | - Xin Qian
- Department of Psychology, Medical School, Jiangsu University, Zhenjiang, China
| | - Fushun Wang
- Institute of Brain and Psychological Sciences, Sichuan Normal University, Chengdu, Sichuan, China
| | - Jason H. Huang
- Department of Neurosurgery, Baylor Scott and White Health, Temple, TX, United States
- Department of Surgery, Texas A&M University, Temple, TX, United States
| |
Collapse
|
13
|
Involvement of the G-Protein-Coupled Estrogen Receptor-1 (GPER) Signaling Pathway in Neurodegenerative Disorders: A Review. Cell Mol Neurobiol 2022:10.1007/s10571-022-01301-9. [DOI: 10.1007/s10571-022-01301-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 10/18/2022] [Indexed: 11/26/2022]
|
14
|
Gohar EY, Almutlaq RN, Fan C, Balkawade RS, Butt MK, Curtis LM. Does G Protein-Coupled Estrogen Receptor 1 Contribute to Cisplatin-Induced Acute Kidney Injury in Male Mice? Int J Mol Sci 2022; 23:ijms23158284. [PMID: 35955435 PMCID: PMC9368456 DOI: 10.3390/ijms23158284] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 07/18/2022] [Accepted: 07/21/2022] [Indexed: 12/04/2022] Open
Abstract
Nephrotoxicity is the dose-limiting side-effect of the chemotherapeutic agent cisplatin (Cp). Recent evidence points to renal protective actions of G protein-coupled estrogen receptor 1 (GPER1). In addition, it has been shown that GPER1 signaling elicits protective actions against acute ischemic injuries that involve multiple organ systems; however, the involvement of GPER1 signaling in Cp-induced acute kidney injury (AKI) remains unclear. This study tested whether genetic deletion of GPER1 exacerbates Cp-induced AKI in male mice. We subjected male mice, homozygous (homo) and heterozygous (het) knockout for the GPER1 gene, and wild-type (WT) littermates to Cp or saline injections and assessed markers for renal injury on the third day after injections. We also determined serum levels of proinflammatory markers in saline and Cp-treated mice. Given the protective role of heme oxygenase-1 (HO-1) in Cp-mediated apoptosis, we also investigated genotypic differences in renal HO-1 abundance, cell death, and proliferation by Western blotting, the TUNEL assay, and Ki67 immunostaining, respectively. Cp increased serum creatinine, urea, and neutrophil gelatinase-associated lipocalin (NGAL) levels, the renal abundance of kidney injury molecule-1, and NGAL in all groups. Cp-induced AKI resulted in comparable histological evidence of injury in all genotypes. WT and homo mice showed greater renal HO-1 abundance in response to Cp. Renal HO-1 abundance was lower in Cp-treated homo, compared to Cp-treated WT mice. Of note, GPER1 deletion elicited a remarkable increase in renal apoptosis; however, no genotypic differences in cell proliferation were observed. Cp augmented kidney Ki67-positive counts, regardless of the genotype. Overall, our data do not support a role for GPER1 in mediating Cp-induced renal injury. GPER1 deletion promotes renal apoptosis and diminishes HO-1 induction in response to Cp, suggesting that GPER1 may play cytoprotective and anti-apoptotic actions in AKI. GPER1-induced regulation of HO-1 and apoptosis may offer novel therapeutic targets for the treatment of AKI.
Collapse
Affiliation(s)
- Eman Y. Gohar
- Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Correspondence: ; Tel.: +1-615-875-0623
| | - Rawan N. Almutlaq
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (R.N.A.); (C.F.); (R.S.B.); (M.K.B.); (L.M.C.)
| | - Chunlan Fan
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (R.N.A.); (C.F.); (R.S.B.); (M.K.B.); (L.M.C.)
| | - Rohan S. Balkawade
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (R.N.A.); (C.F.); (R.S.B.); (M.K.B.); (L.M.C.)
| | - Maryam K. Butt
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (R.N.A.); (C.F.); (R.S.B.); (M.K.B.); (L.M.C.)
| | - Lisa M. Curtis
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (R.N.A.); (C.F.); (R.S.B.); (M.K.B.); (L.M.C.)
| |
Collapse
|
15
|
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
|
16
|
Hirtz A, Lebourdais N, Rech F, Bailly Y, Vaginay A, Smaïl-Tabbone M, Dubois-Pot-Schneider H, Dumond H. GPER Agonist G-1 Disrupts Tubulin Dynamics and Potentiates Temozolomide to Impair Glioblastoma Cell Proliferation. Cells 2021; 10:cells10123438. [PMID: 34943948 PMCID: PMC8699794 DOI: 10.3390/cells10123438] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 11/29/2021] [Accepted: 12/03/2021] [Indexed: 12/11/2022] Open
Abstract
Glioblastoma (GBM) is the most common brain tumor in adults, which is very aggressive, with a very poor prognosis that affects men twice as much as women, suggesting that female hormones (estrogen) play a protective role. With an in silico approach, we highlighted that the expression of the membrane G-protein-coupled estrogen receptor (GPER) had an impact on GBM female patient survival. In this context, we explored for the first time the role of the GPER agonist G-1 on GBM cell proliferation. Our results suggested that G-1 exposure had a cytostatic effect, leading to reversible G2/M arrest, due to tubulin polymerization blockade during mitosis. However, the observed effect was independent of GPER. Interestingly, G-1 potentiated the efficacy of temozolomide, the current standard chemotherapy treatment, since the combination of both treatments led to prolonged mitotic arrest, even in a temozolomide less-sensitive cell line. In conclusion, our results suggested that G-1, in combination with standard chemotherapy, might be a promising way to limit the progression and aggressiveness of GBM.
Collapse
Affiliation(s)
- Alex Hirtz
- Université de Lorraine, CNRS, CRAN, F-54000 Nancy, France; (A.H.); (N.L.); (F.R.); (Y.B.); (A.V.); (H.D.-P.-S.)
| | - Nolwenn Lebourdais
- Université de Lorraine, CNRS, CRAN, F-54000 Nancy, France; (A.H.); (N.L.); (F.R.); (Y.B.); (A.V.); (H.D.-P.-S.)
| | - Fabien Rech
- Université de Lorraine, CNRS, CRAN, F-54000 Nancy, France; (A.H.); (N.L.); (F.R.); (Y.B.); (A.V.); (H.D.-P.-S.)
- Université de Lorraine, CHRU-Nancy, Service de Neurochirurgie, F-54000 Nancy, France
| | - Yann Bailly
- Université de Lorraine, CNRS, CRAN, F-54000 Nancy, France; (A.H.); (N.L.); (F.R.); (Y.B.); (A.V.); (H.D.-P.-S.)
| | - Athénaïs Vaginay
- Université de Lorraine, CNRS, CRAN, F-54000 Nancy, France; (A.H.); (N.L.); (F.R.); (Y.B.); (A.V.); (H.D.-P.-S.)
- Université de Lorraine, CNRS, Inria, LORIA, F-54000 Nancy, France;
| | | | - Hélène Dubois-Pot-Schneider
- Université de Lorraine, CNRS, CRAN, F-54000 Nancy, France; (A.H.); (N.L.); (F.R.); (Y.B.); (A.V.); (H.D.-P.-S.)
| | - Hélène Dumond
- Université de Lorraine, CNRS, CRAN, F-54000 Nancy, France; (A.H.); (N.L.); (F.R.); (Y.B.); (A.V.); (H.D.-P.-S.)
- Correspondence: ; Tel.: +33-372746115
| |
Collapse
|
17
|
Nuclear Receptors in Myocardial and Cerebral Ischemia-Mechanisms of Action and Therapeutic Strategies. Int J Mol Sci 2021; 22:ijms222212326. [PMID: 34830207 PMCID: PMC8617737 DOI: 10.3390/ijms222212326] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 11/08/2021] [Accepted: 11/10/2021] [Indexed: 12/12/2022] Open
Abstract
Nearly 18 million people died from cardiovascular diseases in 2019, of these 85% were due to heart attack and stroke. The available therapies although efficacious, have narrow therapeutic window and long list of contraindications. Therefore, there is still an urgent need to find novel molecular targets that could protect the brain and heart against ischemia without evoking major side effects. Nuclear receptors are one of the promising targets for anti-ischemic drugs. Modulation of estrogen receptors (ERs) and peroxisome proliferator-activated receptors (PPARs) by their ligands is known to exert neuro-, and cardioprotective effects through anti-apoptotic, anti-inflammatory or anti-oxidant action. Recently, it has been shown that the expression of aryl hydrocarbon receptor (AhR) is strongly increased after brain or heart ischemia and evokes an activation of apoptosis or inflammation in injury site. We hypothesize that activation of ERs and PPARs and inhibition of AhR signaling pathways could be a promising strategy to protect the heart and the brain against ischemia. In this Review, we will discuss currently available knowledge on the mechanisms of action of ERs, PPARs and AhR in experimental models of stroke and myocardial infarction and future perspectives to use them as novel targets in cardiovascular diseases.
Collapse
|
18
|
Mechanosensing and the Hippo Pathway in Microglia: A Potential Link to Alzheimer's Disease Pathogenesis? Cells 2021; 10:cells10113144. [PMID: 34831369 PMCID: PMC8622675 DOI: 10.3390/cells10113144] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 10/27/2021] [Accepted: 10/29/2021] [Indexed: 01/01/2023] Open
Abstract
The activation of microglia, the inflammatory cells of the central nervous system (CNS), has been linked to the pathogenesis of Alzheimer’s disease and other neurodegenerative diseases. How microglia sense the changing brain environment, in order to respond appropriately, is still being elucidated. Microglia are able to sense and respond to the mechanical properties of their microenvironment, and the physical and molecular pathways underlying this mechanosensing/mechanotransduction in microglia have recently been investigated. The Hippo pathway functions through mechanosensing and subsequent protein kinase cascades, and is critical for neuronal development and many other cellular processes. In this review, we examine evidence for the potential involvement of Hippo pathway components specifically in microglia in the pathogenesis of Alzheimer’s disease. We suggest that the Hippo pathway is worth investigating as a mechanosensing pathway in microglia, and could be one potential therapeutic target pathway for preventing microglial-induced neurodegeneration in AD.
Collapse
|
19
|
Du ZR, Gu Y, Xie XM, Zhang M, Jiang GY, Chen WF. GPER and IGF-1R mediate the anti-inflammatory effect of genistein against lipopolysaccharide (LPS)-induced nigrostriatal injury in rats. J Steroid Biochem Mol Biol 2021; 214:105989. [PMID: 34478828 DOI: 10.1016/j.jsbmb.2021.105989] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 08/05/2021] [Accepted: 08/25/2021] [Indexed: 12/25/2022]
Abstract
Neuroinflammation plays an important role in the pathogenesis of Parkinson's disease (PD). Genistein is an estrogen-like phytoestrogen that can exert biological effects via the crosstalk of estrogen receptor and insulin-like growth factor 1 receptor (IGF-1R). The present study aimed to evaluate the involvement of G protein-coupled estrogen receptor (GPER) and IGF-1R in the anti-inflammatory effects of genistein against lipopolysaccharide (LPS)-induced nigrostriatal injury in ovariectomized rats. Our results showed that genistein treatment could ameliorate the apomorphine-induced rotational behavior in LPS-induced inflammatory PD rat model. Genistein attenuated LPS-induced decrease of the contents of dopamine (DA) and its metabolites in striatum as well as the loss of tyrosine hydroxylase-immunoreactive (TH-IR) neurons in the substantia nigra (SN) of the lesioned side, which could be blocked by GPER antagonist G15 or IGF-1R antagonist JB1. Meanwhile, G15 or JB1 could attenuate the anti-inflammatory effects of genistein in LPS-induced microglial activation and production of tumor necrosis factor-α (TNF-α), interleukin 1β (IL-1β), inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2). Moreover, genistein could inhibit the LPS-induced phosphorylation of p38, JNK, ERK and IκB in the lesioned side of SN and these effects could also be blocked by G15 or JB1. Taken together, our data provide the first evidence that genistein can inhibit the increase of microglia and protect dopaminergic neurons at least in part via GPER and IGF-1R signaling pathways in ovariectomized PD rat model.
Collapse
Affiliation(s)
- Zhong-Rui Du
- Department of Physiology, Shandong Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao, 266071, China; Laboratory of Sports of Human Science, Ludong University, Yantai, 264000, China
| | - Yu Gu
- Department of Physiology, Shandong Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao, 266071, China
| | - Xiao-Man Xie
- Department of Physiology, Shandong Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao, 266071, China
| | - Mei Zhang
- Department of Physiology, Shandong Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao, 266071, China
| | - Guo-Yi Jiang
- Department of Physiology, Shandong Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao, 266071, China
| | - Wen-Fang Chen
- Department of Physiology, Shandong Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao, 266071, China.
| |
Collapse
|
20
|
From Menopause to Neurodegeneration-Molecular Basis and Potential Therapy. Int J Mol Sci 2021; 22:ijms22168654. [PMID: 34445359 PMCID: PMC8395405 DOI: 10.3390/ijms22168654] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 08/09/2021] [Accepted: 08/09/2021] [Indexed: 12/12/2022] Open
Abstract
The impacts of menopause on neurodegenerative diseases, especially the changes in steroid hormones, have been well described in cell models, animal models, and humans. However, the therapeutic effects of hormone replacement therapy on postmenopausal women with neurodegenerative diseases remain controversial. The steroid hormones, steroid hormone receptors, and downstream signal pathways in the brain change with aging and contribute to disease progression. Estrogen and progesterone are two steroid hormones which decline in circulation and the brain during menopause. Insulin-like growth factor 1 (IGF-1), which plays an import role in neuroprotection, is rapidly decreased in serum after menopause. Here, we summarize the actions of estrogen, progesterone, and IGF-1 and their signaling pathways in the brain. Since the incidence of Alzheimer’s disease (AD) is higher in women than in men, the associations of steroid hormone changes and AD are emphasized. The signaling pathways and cellular mechanisms for how steroid hormones and IGF-1 provide neuroprotection are also addressed. Finally, the molecular mechanisms of potential estrogen modulation on N-methyl-d-aspartic acid receptors (NMDARs) are also addressed. We provide the viewpoint of why hormone therapy has inconclusive results based on signaling pathways considering their complex response to aging and hormone treatments. Nonetheless, while diagnosable AD may not be treatable by hormone therapy, its preceding stage of mild cognitive impairment may very well be treatable by hormone therapy.
Collapse
|
21
|
Network pharmacology identifies IL6 as an important hub and target of tibolone for drug repurposing in traumatic brain injury. Biomed Pharmacother 2021; 140:111769. [PMID: 34058440 DOI: 10.1016/j.biopha.2021.111769] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 05/19/2021] [Accepted: 05/24/2021] [Indexed: 02/06/2023] Open
Abstract
Traumatic brain injury (TBI) is characterized by a complex network of signals mediating inflammatory, proliferative and apoptotic processes during its acute and chronic phases. Current therapies mitigate damage and are mainly for palliative care and there are currently no effective therapies for secondary damage. This suggests a need to discover a compound with a greater spectrum of action that can control various pathological aspects of TBI. Here we used a network pharmacology approach to explore the benefits of tibolone, an estrogen and androgen receptor agonist with broader actions in cells, as a possible repurposing drug for TBI therapy. Using different databases we retrieved the targets significantly associated to TBI and tibolone, obtaining 2700 and 652, respectively. The top 10 GO enriched terms were mostly related to cell proliferation, apoptosis and inflammation. Following protein-protein functional analysis, the top connected proteins were related to kinase activity (MAPK1/14/3, AKT1 PIK3R1), apoptosis (TP53, CASP3), growth factors (EGFR), estrogen signalling (ESR1) and inflammation (IL6, TNF), with IL6 as an important signalling hub belonging to the top GO categories. Thus, we identified IL6 as a cellular node which we then validated using molecular mechanics-generalized born surface area (MMGBSA) and docking to explore which tibolone metabolite might interact with this protein. Both 3α and 3β-OH tibolone seemed to bind better to IL6 at important sites responsible for its binding to IL6R. In conclusion, our study demonstrates key hubs involved in TBI pathology which indicates IL6 as a target molecule of tibolone as drug repurposing for TBI therapy.
Collapse
|
22
|
Gohar EY, Almutlaq RN, Daugherty EM, Butt MK, Jin C, Pollock JS, Pollock DM, De Miguel C. Activation of G protein-coupled estrogen receptor 1 ameliorates proximal tubular injury and proteinuria in Dahl salt-sensitive female rats. Am J Physiol Regul Integr Comp Physiol 2021; 320:R297-R306. [PMID: 33407017 PMCID: PMC7988769 DOI: 10.1152/ajpregu.00267.2020] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 12/21/2020] [Accepted: 12/21/2020] [Indexed: 01/02/2023]
Abstract
Recent evidence indicates a crucial role for G protein-coupled estrogen receptor 1 (GPER1) in the maintenance of cardiovascular and kidney health in females. The current study tested whether GPER1 activation ameliorates hypertension and kidney damage in female Dahl salt-sensitive (SS) rats fed a high-salt (HS) diet. Adult female rats were implanted with telemetry transmitters for monitoring blood pressure and osmotic minipumps releasing G1 (selective GPER1 agonist, 400 μg/kg/day ip) or vehicle. Two weeks after pump implantation, rats were shifted from a normal-salt (NS) diet (0.4% NaCl) to a matched HS diet (4.0% NaCl) for 2 wk. Twenty-four hour urine samples were collected during both diet periods and urinary markers of kidney injury were assessed. Histological assessment of kidney injury was conducted after the 2-wk HS diet period. Compared with values during the NS diet, 24-h mean arterial pressure markedly increased in response to HS, reaching similar values in vehicle-treated and G1-treated rats. HS also significantly increased urinary excretion of protein, albumin, nephrin (podocyte damage marker), and KIM-1 (proximal tubule injury marker) in vehicle-treated rats. Importantly, G1 treatment prevented the HS-induced proteinuria, albuminuria, and increase in KIM-1 excretion but not nephrinuria. Histological analysis revealed that HS-induced glomerular damage did not differ between groups. However, G1 treatment preserved proximal tubule brush-border integrity in HS-fed rats. Collectively, our data suggest that GPER1 activation protects against HS-induced proteinuria and albuminuria in female Dahl SS rats by preserving proximal tubule brush-border integrity in a blood pressure-independent manner.
Collapse
Affiliation(s)
- Eman Y Gohar
- Cardio-Renal Physiology and Medicine Section, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Rawan N Almutlaq
- Cardio-Renal Physiology and Medicine Section, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Elizabeth M Daugherty
- Cardio-Renal Physiology and Medicine Section, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Maryam K Butt
- Cardio-Renal Physiology and Medicine Section, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Chunhua Jin
- Cardio-Renal Physiology and Medicine Section, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Jennifer S Pollock
- Cardio-Renal Physiology and Medicine Section, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - David M Pollock
- Cardio-Renal Physiology and Medicine Section, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Carmen De Miguel
- Cardio-Renal Physiology and Medicine Section, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
23
|
Nguyen DH, Cunningham JT, Sumien N. Estrogen receptor involvement in vascular cognitive impairment and vascular dementia pathogenesis and treatment. GeroScience 2021; 43:159-166. [PMID: 32902819 PMCID: PMC8050128 DOI: 10.1007/s11357-020-00263-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 09/02/2020] [Indexed: 01/26/2023] Open
Abstract
Vascular cognitive impairment (VCI) is a term that encompasses a continuum of cognitive disorders with cerebrovascular pathology contribution, ranging from mild cognitive impairment to vascular dementia (VaD). VCI and VaD, thus, represent an interesting intersection between cardiovascular disease and neurodegenerative disorders such as Alzheimer's disease (AD) and a rising area of research in recent years. Although VCI and VaD research has identified various causes and explanations for disease development, many aspects remain unclear, particularly sex differences in VCI (e.g., epidemiology), unlike those available for cardiovascular disease and AD. Despite limited information in the literature, several studies have observed an association of estrogen receptor (ER) polymorphisms and VaD. If further explored, this association could provide valuable insights for novel therapeutic approaches. This review aims to provide a brief epidemiological overview and subsequent discussion exploring concepts of brain aging and involvement of estrogen receptors in potential mechanisms of VCI/VaD pathogenesis and treatment development.
Collapse
Affiliation(s)
- Dianna H Nguyen
- Department of Physiology and Anatomy, UNT Health Science Center, 3500 Camp Bowie Blvd., Fort Worth, TX, 76107, USA
- Texas College of Osteopathic Medicine, UNT Health Science Center, Fort Worth, TX, USA
| | - J Thomas Cunningham
- Department of Physiology and Anatomy, UNT Health Science Center, 3500 Camp Bowie Blvd., Fort Worth, TX, 76107, USA.
| | - Nathalie Sumien
- Department of Pharmacology and Neuroscience, UNT Health Science Center, Fort Worth, TX, USA
| |
Collapse
|
24
|
Tang Z, Li Q, Cheng Q, Mei M, Song Y, Du Z, He W, Hu J, Yang S, Wang Z. G Protein-Coupled Estrogen Receptor 1 (GPER1) Mediates Aldosterone-Induced Endothelial Inflammation in a Mineralocorticoid Receptor-Independent Manner. Int J Endocrinol 2021; 2021:5575927. [PMID: 34239558 PMCID: PMC8235990 DOI: 10.1155/2021/5575927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 06/04/2021] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVE It has been increasingly appreciated that G protein-coupled estrogen receptor 1 (GPER1) mediates both proinflammatory and anti-inflammatory response of estrogen. It is also involved in some rapid vascular effects of aldosterone in a mineralocorticoid receptor (MR) independent manner. However, whether GPER1 mediates aldosterone-induced inflammation response in endothelial cells and its relationship with MR are yet undetermined and therefore require further explanation. METHOD Based on the hypothesis that GPER1 plays a role in the aldosterone-related vascular inflammation, the present study utilized a model of human umbilical vein endothelial cells transfected with MR siRNA and induced for inflammatory response with increasing concentration of aldosterone. RESULTS It was discovered that induction of aldosterone had no effect on the expression of GPER1 but promoted the expression of MR. Suppression of MR did not influence GPER1 expression, and GPER1 was capable of mediating part of aldosterone-induced endothelial inflammatory response. This effect may involve phosphoinositide 3-kinases (PI3K) pathway signaling. CONCLUSION These findings not only demonstrated the role of GPER1 in aldosterone-induced vascular inflammation but also suggested an alternative for pharmaceutical treatment of hyperaldosteronism considering the unsatisfying effect on cardiovascular risks with MR antagonists.
Collapse
Affiliation(s)
- Ziwei Tang
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400044, China
| | - Qifu Li
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400044, China
| | - Qingfeng Cheng
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400044, China
| | - Mei Mei
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400044, China
| | - Ying Song
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400044, China
| | - Zhipeng Du
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400044, China
| | - Wenwen He
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400044, China
| | - Jinbo Hu
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400044, China
| | - Shumin Yang
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400044, China
| | - Zhihong Wang
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400044, China
| |
Collapse
|
25
|
Gagne C, Piot A, Brake WG. Depression, Estrogens, and Neuroinflammation: A Preclinical Review of Ketamine Treatment for Mood Disorders in Women. Front Psychiatry 2021; 12:797577. [PMID: 35115970 PMCID: PMC8804176 DOI: 10.3389/fpsyt.2021.797577] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 12/24/2021] [Indexed: 12/11/2022] Open
Abstract
Ketamine has been shown to acutely and rapidly ameliorate depression symptoms and suicidality. Given that women suffer from major depression at twice the rate of men, it is important to understand how ketamine works in the female brain. This review explores three themes. First, it examines our current understanding of the etiology of depression in women. Second, it examines preclinical research on ketamine's antidepressant effects at a neurobiological level as well as how ovarian hormones present a unique challenge in interpreting these findings. Lastly, the neuroinflammatory hypothesis of depression is highlighted to help better understand how ovarian hormones might interact with ketamine in the female brain.
Collapse
Affiliation(s)
- Collin Gagne
- Department of Psychology, Centre for Studies in Behavioural Neurobiology Concordia University, Montreal, QC, Canada
| | - Alexandre Piot
- Department of Psychology, Centre for Studies in Behavioural Neurobiology Concordia University, Montreal, QC, Canada
| | - Wayne G Brake
- Department of Psychology, Centre for Studies in Behavioural Neurobiology Concordia University, Montreal, QC, Canada
| |
Collapse
|
26
|
Wang T, Chen H, Xia S, Chen X, Sun H, Xu Z. Ameliorative Effect of Parishin C Against Cerebral Ischemia-Induced Brain Tissue Injury by Reducing Oxidative Stress and Inflammatory Responses in Rat Model. Neuropsychiatr Dis Treat 2021; 17:1811-1823. [PMID: 34113111 PMCID: PMC8187103 DOI: 10.2147/ndt.s309065] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 05/11/2021] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Gastrodia elata Blume (Orchidaceae) is a widely used traditional Chinese herbal medicine in the clinical practice of China, to treat nervous headache, convulsions, dizziness, neurasthenia, and so on. Parishin C (Par C), one of the major bioactive components of Gastrodia elata Blume, is known to exert many different biological activities, including antipsychotic and neuroprotective effects. However, there is little research about its neuroprotective effect in an ischemic stroke model. The objective of the present study is thus to investigate the neuroprotective effects of Par C against cerebral ischemia damage. METHODS Rats were pretreated with Par C (25, 50, or 100 mg/kg/day, i.p.) for 21 days, then subjected to 2 h of middle cerebral artery occlusion (MCAO) and 22 h of reperfusion. Neurological deficient scores, brain water content, histopathology, TCC staining were performed to assess the neuroprotective effects of Par C. Meanwhile, the oxidative stress, inflammation and apoptosis-related markers of brain tissue were evaluated by corresponding assay kits. Besides, the antioxidant and pro-inflammatory expression was measured by real-time quantification PCR (RT-qPCR). RESULTS Our findings indicated that the pre-treatment with Par C improved nerve function, suppressed oxidative stress, and pro-inflammatory factors release in rats with cerebral ischemia damage. Besides, Par C significantly increased antioxidant expression and declined pro-inflammatory cytokines expression. CONCLUSION Par C is shown to exert neuroprotective effects partly via inhibiting oxidative stress and inflammation in a rat model of MCAO.
Collapse
Affiliation(s)
- Tao Wang
- Department of Anesthesiology, the Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan, 570311, People's Republic of China
| | - Haibo Chen
- Department of Blood Transfusion, the Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan, 570311, People's Republic of China
| | - Shuyun Xia
- Department of Respiratory Medicine, Pingdu People's Hospital, Pingdu, Shandong, 266700, People's Republic of China
| | - Xiaofang Chen
- Department of Anesthesiology, the Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan, 570311, People's Republic of China
| | - Hu Sun
- Department of Anesthesiology, the Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan, 570311, People's Republic of China
| | - Zhixin Xu
- Department of Anesthesiology, the Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan, 570311, People's Republic of China
| |
Collapse
|
27
|
Sen A, Kaul A, Kaul R. Estrogen receptors in human bladder cells regulate innate cytokine responses to differentially modulate uropathogenic E. coli colonization. Immunobiology 2020; 226:152020. [PMID: 33246308 DOI: 10.1016/j.imbio.2020.152020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 08/19/2020] [Accepted: 10/18/2020] [Indexed: 10/23/2022]
Abstract
The bladder epithelial cells elicit robust innate immune responses against urinary tract infections (UTIs) for preventing the bacterial colonization. Physiological fluctuations in circulating estrogen levels in women increase the susceptibility to UTI pathogenesis, often resulting in adverse health outcomes. Dr adhesin bearing Escherichia coli (Dr E. coli) cause recurrent UTIs in menopausal women and acute pyelonephritis in pregnant women. Dr E. coli bind to epithelial cells via host innate immune receptor CD55, under hormonal influence. The role of estrogens or estrogen receptors (ERs) in regulating the innate immune responses in the bladder are poorly understood. In the current study, we investigated the role of ERα, ERβ and GPR30 in modulating the innate immune responses against Dr E. coli induced UTI using human bladder epithelial carcinoma 5637 cells (HBEC). Both ERα and ERβ agonist treatment in bladder cells induced a protection against Dr E. coli invasion via upregulation of TNFα and downregulation of CD55 and IL10, and these effects were reversed by action of ERα and ERβ antagoinsts. In contrast, the agonist-mediated activation of GPR30 led to an increased bacterial colonization due to suppression of innate immune factors in the bladder cells, and these effects were reversed by the antagonist-mediated suppression of GPR30. Further, siRNA-mediated ERα knockdown in the bladder cells reversed the protection against bacterial invasion observed in the ERα positive bladder cells, by modulating the gene expression of TNFα, CD55 and IL10, thus confirming the protective role of ERα. We demonstrate for the first time a protective role of nuclear ERs, ERα and ERβ but not of membrane ER, GPR30 against Dr E. coli invasion in HBEC 5637 cells. These findings have many clinical implications and suggest that ERs may serve as potential drug targets towards developing novel therapeutics for regulating local innate immunity and treating UTIs.
Collapse
Affiliation(s)
- Ayantika Sen
- Department of Biochemistry and Microbiology, Oklahoma State University Center for Health Sciences, 1111 West 17th Street, Tulsa, OK 74107, USA; Division of Abdominal Transplantation, Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Anil Kaul
- Health Care Administration, Oklahoma State University Center for Health Sciences, 1111 West 17th Street, Tulsa, OK 74107, USA
| | - Rashmi Kaul
- Department of Biochemistry and Microbiology, Oklahoma State University Center for Health Sciences, 1111 West 17th Street, Tulsa, OK 74107, USA.
| |
Collapse
|
28
|
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: 7] [Impact Index Per Article: 1.8] [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
|
29
|
Vahidinia Z, Karimian M, Joghataei MT. Neurosteroids and their receptors in ischemic stroke: From molecular mechanisms to therapeutic opportunities. Pharmacol Res 2020; 160:105163. [DOI: 10.1016/j.phrs.2020.105163] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 08/17/2020] [Accepted: 08/17/2020] [Indexed: 01/09/2023]
|
30
|
Erfani S, Valadbeigi T, Aboutaleb N, Karimi N, Moghimi A, Khaksari M. Usnic acid improves memory impairment after cerebral ischemia/reperfusion injuries by anti-neuroinflammatory, anti-oxidant, and anti-apoptotic properties. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2020; 23:1225-1231. [PMID: 32963745 PMCID: PMC7491502 DOI: 10.22038/ijbms.2020.43280.10165] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Objective(s): Cerebral ischemia/reperfusion causes complex pathological mechanisms that lead to brain tissue damage. Usnic acid is a lichen secondary metabolite that has many different biological properties including anti-inflammatory and anti-oxidant activities. Therefore, the objective of the current study was to investigate the neuroprotective effects of usnic acid on apoptotic cell death, neuroinflammation, anti-oxidant enzyme activities, and oxidative stress levels after transient cerebral ischemia/reperfusion. Materials and Methods: Forty-two male Wistar rats were randomly assigned to three groups (sham, ischemia/reperfusion, and ischemia/reperfusion+usnic acid). Ischemia was induced by 20 min occlusion of common carotid arteries. Injection of usnic acid (25 mg/kg, intraperitoneally) and saline was done at the beginning of reperfusion time. Morris water maze was applied to assess spatial memory. The protein expression amount was measured using immunohistochemical and immunofluorescence staining. Spectrophotometric assay was performed to determine the levels of anti-oxidant enzymes. Results: Usnic acid significantly reduced caspase-3, glial fibrillary acidic protein- positive and ionized calcium-binding adaptor molecule 1-positive cells (P<0.001) and enhanced spatial memory disorders (P<0.05) due to brain ischemia. In addition, treatment with usnic acid improves effects in the antioxidant system following cerebral ischemia (P<0.05). Conclusion: Our findings indicate that usnic acid has neuroprotective properties, which possibly is applicable as a promising candidate for cerebral injuries caused by ischemia.
Collapse
Affiliation(s)
- Sohaila Erfani
- Department of Biology, Faculty of Science, Ilam University, Ilam, Iran
| | | | - Nahid Aboutaleb
- Physiology Research Center and Department of Physiology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran.,Department of Physiology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Naser Karimi
- Department of Biology, Faculty of Science, Razi University of Kermanshah, Kermanshah, Iran
| | - Ali Moghimi
- Rayan Center for Neuroscience and Behavior, Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Mehdi Khaksari
- School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| |
Collapse
|
31
|
Pan MX, Li J, Ma C, Fu K, Li ZQ, Wang ZF. Sex-dependent effects of GPER activation on neuroinflammation in a rat model of traumatic brain injury. Brain Behav Immun 2020; 88:421-431. [PMID: 32272225 DOI: 10.1016/j.bbi.2020.04.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 04/03/2020] [Accepted: 04/04/2020] [Indexed: 02/06/2023] Open
Abstract
The G protein-coupled estrogen receptor (GPER) plays a role in estrogen-mediated neuroprotection and has been considered a potential therapeutic target for treating various neurological diseases. It is increasingly recognized that sex is a biological variable affecting treatment outcomes and efficacy, and that neuroinflammation is a key secondary injury mechanism following brain injury, though it is unknown whether the neuroprotective effects exerted by GPER involve modulation of inflammatory processes. The aim of this study was to investigate whether activation of GPER has a sex-dependent effect on neuroinflammation following traumatic brain injury (TBI), a sexually dimorphic disease. In male and ovariectomized (OVX) female rats, the GPER agonist, G1, inhibited the upregulated expression of pro-inflammatory cytokines (IL-1β, IL-6, and TNF-α), increased the expression of the anti-inflammatory cytokine IL-4, and shifted microglia/macrophage polarization toward the M2 phenotype. In gonadally-intact females, G1 caused more pro-inflammatory (IL-6 and TNF-α) and less anti-inflammatory cytokine (IL-4) production, without altering microglia/macrophage polarization. Estradiol supplementation blocked the effects of G1 in OVX females. We also found that post-injury GPER expression was increased in males and OVX females but not in intact females. G1 administration increased Akt phosphorylation in males and OVX females, but had no significant effect in intact females, while Akt inhibition blocked the effects of G1 in males and OVX females. These results indicate that G1 exerts anti-inflammatory effects in males and OVX females but not in intact females; these sex-specific effects are dependent on circulating estrogen levels and are partially mediated through Akt signaling. Future studies are needed to elucidate the relevant molecular mechanisms, especially in females. A better understanding of the sex differences in treatment efficacy with GPER agonists may help improve personalized therapeutic strategies for males and pre- and postmenopausal females with TBI.
Collapse
Affiliation(s)
- Meng-Xian Pan
- Department of Physiology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| | - Jie Li
- Department of Physiology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| | - Chao Ma
- Department of Neurosurgery, Wuhan University Zhongnan Hospital, Wuhan 430071, China
| | - Kai Fu
- Department of Neurosurgery, Wuhan University Zhongnan Hospital, Wuhan 430071, China
| | - Zhi-Qiang Li
- Department of Neurosurgery, Wuhan University Zhongnan Hospital, Wuhan 430071, China.
| | - Ze-Fen Wang
- Department of Physiology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China.
| |
Collapse
|
32
|
Yao YY, Ling EA, Lu D. Microglia mediated neuroinflammation - signaling regulation and therapeutic considerations with special reference to some natural compounds. Histol Histopathol 2020; 35:1229-1250. [PMID: 32662061 DOI: 10.14670/hh-18-239] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Neuroinflammation plays a central role in multiple neurodegenerative diseases and neurological disorders such as Alzheimer's disease (AD), Parkinson's disease (PD), cerebral ischemic injury etc. In this connection, microglia, the key players in the central nervous system, mediate the inflammatory response process. In brain injuries, activated microglia can clear the cellular debris and invading pathogens and release neurotrophic factors; however, prolonged microglia activation may cause neuronal death through excessive release of inflammatory mediators. Therefore, it is of paramount importance to understand the underlying molecular mechanisms of microglia activation to design an effective therapeutic strategy to alleviate neuronal injury. Recent studies have shown that some natural compounds and herbal extracts possess anti-inflammatory properties that may suppress microglial activation and ameliorate neuroinflammation and hence are neuroprotective. In this review, we will update some of the common signaling pathways that regulate microglia activation. Among the various signaling pathways, the Notch-1, mitogen-activated protein kinases (MAPKs), and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) have been reported to exacerbate microglia mediated neuroinflammation that is implicated in different neuropathological diseases. The search for natural compounds or agents, specifically those derived from natural herbal extracts such as Gastrodin, scutellarin, RG1 etc. has been the focus of many of our recent studies because they have been found to regulate microglia activation. The pharmacological effects of these agents and their potential mechanisms for regulating microglia activation are systematically reviewed here for a fuller understanding of their biochemical action and therapeutic potential for treatment of microglia mediated neuropathological diseases.
Collapse
Affiliation(s)
- Yue-Yi Yao
- Technology Transfer Center, Kunming Medical University, Kunming, China
| | - Eng-Ang Ling
- Department of Anatomy, Young Loo Lin School of Medicine, National University of Singapore, Singapore.
| | - Di Lu
- Technology Transfer Center, Kunming Medical University, Kunming, China.
| |
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. [DOI: 10.1124/jpet.119.264598] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 05/05/2020] [Indexed: 12/16/2022] Open
|
34
|
Microglial and Astrocytic Function in Physiological and Pathological Conditions: Estrogenic Modulation. Int J Mol Sci 2020; 21:ijms21093219. [PMID: 32370112 PMCID: PMC7247358 DOI: 10.3390/ijms21093219] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 04/24/2020] [Accepted: 04/30/2020] [Indexed: 12/20/2022] Open
Abstract
There are sexual differences in the onset, prevalence, and outcome of numerous neurological diseases. Thus, in Alzheimer’s disease, multiple sclerosis, and major depression disorder, the incidence in women is higher than in men. In contrast, men are more likely to present other pathologies, such as amyotrophic lateral sclerosis, Parkinson’s disease, and autism spectrum. Although the neurological contribution to these diseases has classically always been studied, the truth is that neurons are not the only cells to be affected, and there are other cells, such as glial cells, that are also involved and could be key to understanding the development of these pathologies. Sexual differences exist not only in pathology but also in physiological processes, which shows how cells are differentially regulated in males and females. One of the reasons these sexual differences may occur could be due to the different action of sex hormones. Many studies have shown an increase in aromatase levels in the brain, which could indicate the main role of estrogens in modulating proinflammatory processes. This review will highlight data about sex differences in glial physiology and how estrogenic compounds, such as estradiol and tibolone, could be used as treatment in neurological diseases due to their anti-inflammatory effects and the ability to modulate glial cell functions.
Collapse
|
35
|
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: 2.5] [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
|
36
|
Deems NP, Leuner B. Pregnancy, postpartum and parity: Resilience and vulnerability in brain health and disease. Front Neuroendocrinol 2020; 57:100820. [PMID: 31987814 PMCID: PMC7225072 DOI: 10.1016/j.yfrne.2020.100820] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 11/25/2019] [Accepted: 01/21/2020] [Indexed: 02/07/2023]
Abstract
Risk and resilience in brain health and disease can be influenced by a variety of factors. While there is a growing appreciation to consider sex as one of these factors, far less attention has been paid to sex-specific variables that may differentially impact females such as pregnancy and reproductive history. In this review, we focus on nervous system disorders which show a female bias and for which there is data from basic research and clinical studies pointing to modification in disease risk and progression during pregnancy, postpartum and/or as a result of parity: multiple sclerosis (MS), depression, stroke, and Alzheimer's disease (AD). In doing so, we join others (Shors, 2016; Galea et al., 2018a) in aiming to illustrate the importance of looking beyond sex in neuroscience research.
Collapse
Affiliation(s)
- Nicholas P Deems
- The Ohio State University, Department of Psychology, Columbus, OH, USA
| | - Benedetta Leuner
- The Ohio State University, Department of Psychology, Columbus, OH, USA.
| |
Collapse
|
37
|
Butler MJ, Perrini AA, Eckel LA. Estradiol treatment attenuates high fat diet-induced microgliosis in ovariectomized rats. Horm Behav 2020; 120:104675. [PMID: 31923417 PMCID: PMC7117977 DOI: 10.1016/j.yhbeh.2020.104675] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Revised: 11/20/2019] [Accepted: 12/28/2019] [Indexed: 01/03/2023]
Abstract
Consumption of a high fat diet (HFD) increases circulating free fatty acids, which can enter the brain and promote a state of microgliosis, as defined by a change in microglia number and/or morphology. Most studies investigating diet-induced microgliosis have been conducted in male rodents despite well-documented sex differences in the neural control of food intake and neuroimmune signaling. This highlights the need to investigate how sex hormones may modulate the behavioral and cellular response to HFD consumption. Estradiol is of particular interest since it exerts a potent anorexigenic effect and has both anti-inflammatory and neuroprotective effects in the brain. As such, the aim of the current study was to investigate whether estradiol attenuates the development of HFD-induced microgliosis in female rats. Estradiol- and vehicle-treated ovariectomized rats were fed either a low-fat chow diet or a 60% HFD for 4 days, after which they were perfused and brain sections were processed via immunohistochemistry for microglia-specific Iba1 protein. Four days of HFD consumption promoted microgliosis, as measured via an increase in the number of microglia in the arcuate nucleus (ARC) of the hypothalamus and nucleus of the solitary tract (NTS), and a decrease in microglial branching in the ARC, NTS, lateral hypothalamus (LH), and ventromedial hypothalamus. Estradiol replacement attenuated the HFD-induced changes in microglia accumulation and morphology in the ARC, LH, and NTS. We conclude that estradiol has protective effects against HFD-induced microgliosis in a region-specific manner in hypothalamic and hindbrain areas implicated in the neural control of food intake.
Collapse
Affiliation(s)
- Michael J Butler
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, FL 32306, United States; Institute for Behavioral Medicine Research, Ohio State University Wexner Medical Center, Columbus, OH, 43210, United States
| | - Alexis A Perrini
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, FL 32306, United States
| | - Lisa A Eckel
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, FL 32306, United States.
| |
Collapse
|
38
|
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: 43] [Impact Index Per Article: 10.8] [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
|
39
|
Activation of the G Protein-Coupled Estrogen Receptor (GPER) Increases Neurogenesis and Ameliorates Neuroinflammation in the Hippocampus of Male Spontaneously Hypertensive Rats. Cell Mol Neurobiol 2019; 40:711-723. [DOI: 10.1007/s10571-019-00766-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 11/22/2019] [Indexed: 01/20/2023]
|
40
|
Gao XQ, Du ZR, Yuan LJ, Zhang WD, Chen L, Teng JJ, Wong MS, Xie JX, Chen WF. Ginsenoside Rg1 Exerts Anti-inflammatory Effects via G Protein-Coupled Estrogen Receptor in Lipopolysaccharide-Induced Microglia Activation. Front Neurosci 2019; 13:1168. [PMID: 31787867 PMCID: PMC6853999 DOI: 10.3389/fnins.2019.01168] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 10/16/2019] [Indexed: 11/13/2022] Open
Abstract
Neuroinflammation plays a pivotal role in the pathogenesis of Parkinson's disease. Ginsenoside Rg1, the most active ingredient of ginseng, has been reported to exert neuroprotective effects via estrogen and glucocorticoid receptors. The present study evaluated the involvement of the G protein-coupled estrogen receptor (GPER) in the anti-inflammatory effects of ginsenoside Rg1 against lipopolysaccharide (LPS)-induced microglia activation in the BV2 microglial cell line and ventral mesencephalic primary microglial culture. The pharmacological blockade and lentivirus-mediated small interfering RNA (siRNA) knockdown of GPER were used to study the underlying mechanism. Rg1 attenuated LPS-induced upregulation of tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), inducible nitric oxide synthase (iNOS), and cyclooxygenase-2 (COX-2) mRNA and protein levels. The GPER antagonist G15 blocked the inhibitory effects of Rg1 and the GPER-specific agonist G1 on LPS-induced microglia activation. Rg1 mimicked the effects of G1 by inhibiting the LPS-induced activation of nuclear transcription factor-kappa B (NF-κB) and mitogen activated protein kinase signaling pathways, which was also blocked by G15. Moreover, lentivirus-mediated siRNA knockdown of GPER inhibited the anti-inflammatory effects of Rg1. Taken together, our results indicate that GPER is involved in the anti-inflammatory effects of Rg1 against LPS-induced microglia activation. These findings provide a new biological target of Rg1 for the treatment of neuroinflammatory disorders.
Collapse
Affiliation(s)
- Xian-Qi Gao
- Department of Physiology, Shandong Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Zhong-Rui Du
- Department of Physiology, Shandong Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Liang-Jie Yuan
- Department of Physiology, Shandong Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao, China.,School of Basic Medicine, Shandong First Medical University and Shandong Academy of Medical Sciences, Tai'an, China
| | - Wen-Di Zhang
- Department of Physiology, Shandong Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Lei Chen
- Department of Physiology, Shandong Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Ji-Jun Teng
- Department of Neurology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Man-Sau Wong
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, China
| | - Jun-Xia Xie
- Department of Physiology, Shandong Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Wen-Fang Chen
- Department of Physiology, Shandong Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao, China
| |
Collapse
|
41
|
Peng J, Zuo Y, Huang L, Okada T, Liu S, Zuo G, Zhang G, Tang J, Xia Y, Zhang JH. Activation of GPR30 with G1 attenuates neuronal apoptosis via src/EGFR/stat3 signaling pathway after subarachnoid hemorrhage in male rats. Exp Neurol 2019; 320:113008. [DOI: 10.1016/j.expneurol.2019.113008] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 05/16/2019] [Accepted: 07/05/2019] [Indexed: 12/21/2022]
|
42
|
Machado GDB, de Freitas BS, Florian LZ, Monteiro RT, Gus H, Schröder N. G protein-coupled oestrogen receptor stimulation ameliorates iron- and ovariectomy-induced memory impairments through the cAMP/PKA/CREB signalling pathway. J Neuroendocrinol 2019; 31:e12780. [PMID: 31418949 DOI: 10.1111/jne.12780] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 07/09/2019] [Accepted: 08/13/2019] [Indexed: 12/18/2022]
Abstract
Iron accumulation in the brain has been associated with neurodegenerative disorders, and imaging studies in humans indicate that iron content in brain regions correlates with poor performance in cognitive tasks. In rats, iron overload impairs memory retention in a variety of memory tasks. Although the effects of iron on cognition in rodents are extensively reported, no previous study has been conducted in female rats. The incidence of certain dementias, such as Alzheimer's disease, is higher in women after menopause compared to aged-matched men. The role of oestrogen depletion in memory deficits in menopausal women is still a matter of debate. The present study aimed to characterise the effects of iron overload on memory in female rats by investigating the effects of ovariectomy (OVX, an experimental model of oestrogen depletion) in rats submitted to iron overload, as well as examining the effects of G protein-coupled oestrogen receptor (GPER) agonism on memory impairments induced by iron and OVX. Female rats received iron (30 mg kg-1 , orally) or vehicle at postnatal days 12-14 and were submitted to OVX in adulthood. Results showed that either iron or OVX impaired memory for object placement and inhibitory avoidance. The selective GPER agonist G1, administered immediately after training, reversed both iron- and OVX-induced memory impairments. G1 effects were abolished by protein kinase A (PKA) inhibition, suggesting the involvement of the cAMP/PKA/CREB signalling pathway. The search for novel oestrogen agonists with positive effects on cognition may be promising for the development of treatments for memory disorders.
Collapse
Affiliation(s)
- Gustavo Dalto Barroso Machado
- Neurobiology and Developmental Biology Laboratory, School of Sciences, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Betânia Souza de Freitas
- Neurobiology and Developmental Biology Laboratory, School of Sciences, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Leonardo Zanetti Florian
- Neurobiology and Developmental Biology Laboratory, School of Sciences, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Ricardo Tavares Monteiro
- Neurobiology and Developmental Biology Laboratory, School of Sciences, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Henrique Gus
- Neurobiology and Developmental Biology Laboratory, School of Sciences, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Nadja Schröder
- National Institute of Science and Technology for Translational Medicine (INCT-TM), Conselho Nacional de Desenvolvimento Cientifico e Tecnologico (CNPq), Brasília, Brazil
- Department of Physiology, Institute for Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|
43
|
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: 2.0] [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
|
44
|
Kim T, Chelluboina B, Chokkalla AK, Vemuganti R. Age and sex differences in the pathophysiology of acute CNS injury. Neurochem Int 2019; 127:22-28. [PMID: 30654116 PMCID: PMC6579702 DOI: 10.1016/j.neuint.2019.01.012] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 01/11/2019] [Indexed: 12/21/2022]
Abstract
Despite the immeasurable burden on patients and families, no effective therapies to protect the CNS after an acute injury are available yet. Furthermore, the underlying mechanisms that promote neuronal death and functional deficits after injury remain to be poorly understood. The prevalence, age of onset, pathophysiology, and symptomatology of many CNS insults differ significantly between males and females. In the case of stroke, younger males tend to show a higher risk than younger females, while this trend reverses with age. Accumulating evidence from preclinical studies have shown that sex hormones play a crucial role in providing neuroprotection following ischemic stroke and other acute CNS injuries. Estrogen, in particular, exerts a neuroprotective effect by modulating the immune responses after injury. In addition, there exists a sexual dimorphism in cell death pathways between males and females that are independent of hormones. Meanwhile, recent studies suggest that microRNAs are critically involved in the sex-specific mechanisms of cell death. This review discusses the current knowledge on the contribution of sex and age to outcome after stroke. Implication of the interplay between these two factors on other CNS injuries (spinal cord injury and traumatic brain injury) from the experimental evidence were also discussed.
Collapse
Affiliation(s)
- TaeHee Kim
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | - Bharath Chelluboina
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | - Anil K Chokkalla
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA; Cellular and Molecular Pathology Program, University of Wisconsin, Madison, WI, USA
| | - Raghu Vemuganti
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA; Cellular and Molecular Pathology Program, University of Wisconsin, Madison, WI, USA; William S. Middleton Veterans Administration Hospital, Madison, WI, USA.
| |
Collapse
|
45
|
Jacenik D, Cygankiewicz AI, Mokrowiecka A, Małecka-Panas E, Fichna J, Krajewska WM. Sex- and Age-Related Estrogen Signaling Alteration in Inflammatory Bowel Diseases: Modulatory Role of Estrogen Receptors. Int J Mol Sci 2019; 20:ijms20133175. [PMID: 31261736 PMCID: PMC6651503 DOI: 10.3390/ijms20133175] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 06/26/2019] [Indexed: 02/07/2023] Open
Abstract
The pathogenesis of inflammatory bowel diseases (IBD) seems to be associated with alterations of immunoregulation. Several lines of evidence suggest that estrogens play a role in the modulation of immune responses and may be related to the etiology of IBD. The purpose of this work was to examine the involvement of G protein-coupled estrogen receptor (GPER), estrogen receptor α (ERα), estrogen receptor β (ERβ) and ERα spliced variants ERα36 and ERα46 in Crohn's disease (CD) and ulcerative colitis (UC). The studied group included 73 patients with IBD and 31 sex and age-related controls. No differences in serum levels of 17β-estradiol nor of CYP1A1 and SULT1E1 enzymes involved in estrogen catabolism were stated. The expression pattern of estrogen receptors in tissue samples was quantified using real-time PCR and Western blotting. Statistically significant up-regulation of GPER and ERα in both CD and UC as well as down-regulation of ERβ in CD patients was found. However, differences in the expression of estrogen receptors in CD and UC have been identified, depending on the sex and age of patients. In men, up-regulation of GPER, ERα and ERα46 expression was shown in CD and UC patients. In women under 50 years of age, GPER protein level increased in UC whereas ERβ expression tended to decrease in CD and UC patients. In turn, in women over 50 the protein level of ERα increased in UC while ERβ expression decreased in CD patients. Dysregulation of estrogen receptors in the intestinal mucosa of patients with CD and UC indicates that estrogen signaling may play a role in the local immune response and maintain epithelial homeostasis in a gender- and age-dependent manner.
Collapse
Affiliation(s)
- Damian Jacenik
- Department of Cytobiochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska St. 141/143, 90-236 Lodz, Poland.
| | - Adam I Cygankiewicz
- Department of Cytobiochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska St. 141/143, 90-236 Lodz, Poland
| | - Anna Mokrowiecka
- Department of Digestive Tract Diseases, Faculty of Medicine, Medical University of Lodz, Stefana Kopcinskiego St. 22, 90-001 Lodz, Poland
| | - Ewa Małecka-Panas
- Department of Digestive Tract Diseases, Faculty of Medicine, Medical University of Lodz, Stefana Kopcinskiego St. 22, 90-001 Lodz, Poland
| | - Jakub Fichna
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Mazowiecka St. 6/8, 92-215 Lodz, Poland
| | - Wanda M Krajewska
- Department of Cytobiochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska St. 141/143, 90-236 Lodz, Poland.
| |
Collapse
|
46
|
Xu F, Shen G, Su Z, He Z, Yuan L. Glibenclamide ameliorates the disrupted blood-brain barrier in experimental intracerebral hemorrhage by inhibiting the activation of NLRP3 inflammasome. Brain Behav 2019; 9:e01254. [PMID: 30859754 PMCID: PMC6456786 DOI: 10.1002/brb3.1254] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 12/15/2018] [Accepted: 02/09/2019] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND Glibenclamide is a widely used sulfonylurea drug prescribed to treat type II diabetes mellitus. Previous studies have demonstrated that glibenclamide has neuroprotective effects in central nervous system injury. However, the exact mechanism by which glibenclamide acts on the blood-brain barrier (BBB) after intracerebral hemorrhage (ICH) remains unclear. The purpose of this study was to validate the neuroprotective effects of glibenclamide on ICH and to explore the mechanisms underlying these effects. METHODS We investigated the effects of glibenclamide on experimental ICH using the autologous blood infusion model. Glibenclamide was administrated either immediately or 2 hr after ICH. Brain edema was quantified using the wet-dry method 3 days after injury. BBB integrity was evaluated by Evans Blue extravasation and degradation of the tight junction protein zona occludens-1 (ZO-1). mRNA levels of inflammatory cytokines were determined by quantitative polymerase chain reaction. Activation of the nucleotide-binding oligomerization domain-like receptor with a pyrin domain 3 (NLRP3) inflammasome and cell viability were also measured in cerebral microvascular endothelial b.End3 cells exposed to hemin. Neurological changes were evaluated by the Garcia score and rotarod test. RESULTS After ICH, the brain water content, Evans Blue extravasation, and inflammatory cytokines decreased significantly in the ipsilateral hemisphere of the experimental compared to the vehicle group. Glibenclamide treatment and NLRP3 knockdown significantly reduced hemin-induced activation of the NLRP3 inflammasome, release of extracellular lactate dehydrogenase, apoptosis, and loss of ZO-1 in b.End3 cells. However, NLRP3 knockdown abolished the protective effect of glibenclamide. CONCLUSION Glibenclamide maintained BBB integrity in experimental ICH by inhibiting the activation of the NLRP3 inflammasome in microvessel endothelial cells. Our findings will contribute to elucidating the pharmacological mechanism of action of glibenclamide and to developing a novel therapy for clinical ICH.
Collapse
Affiliation(s)
- Fulin Xu
- Department of Neurosurgery, Minhang District Central hospital, Shanghai, China
| | - Gang Shen
- Department of Neurosurgery, Minhang District Central hospital, Shanghai, China
| | - Zuopeng Su
- Department of Neurosurgery, Minhang District Central hospital, Shanghai, China
| | - Zijian He
- Department of Neurosurgery, Minhang District Central hospital, Shanghai, China
| | - Lutao Yuan
- Department of Neurosurgery, Minhang District Central hospital, Shanghai, China
| |
Collapse
|
47
|
Erfani S, Moghimi A, Aboutaleb N, Khaksari M. Protective effects of Nesfatin-1 peptide on cerebral ischemia reperfusion injury via inhibition of neuronal cell death and enhancement of antioxidant defenses. Metab Brain Dis 2019; 34:79-85. [PMID: 30269302 DOI: 10.1007/s11011-018-0323-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 09/24/2018] [Indexed: 01/04/2023]
Abstract
Nesfatin-1 is a novel peptide with anorexigenic and anti-hyperglycemic properties. According to previous studies, this multi-functional peptide protects dopaminergic cells against neurotoxicity via anti-apoptotic effects. In addition, Nesfatin-1 protects myocardial tissue after myocardial infarction via anti-inflammatory and anti-apoptotic mechanisms. In this study, we investigated the neuroprotective effects of nesfatin-1 against cerebral ischemia reperfusion injury in the CA1 area of hippocampus in rats. 56 male Wistar rats (240-270 g) were randomly selected and allocated into four groups: (1) sham, (2) nesfatin-1, (3) ischemia/reperfusion, (4) ischemia/reperfusion+nesfatin-1. Cerebral ischemia induced by the occlusion of the common carotid arteries for 20 min was followed by reperfusion. Saline as a vehicle and nesfatin-1 (20 μg/kg) were injected intraperitoneally (IP) at the start of cerebral reperfusion. Apoptotic and necrotic cell death was detected by TUNEL and Nissl staining. Malondialdehyde (MDA) and antioxidant enzymes (GSH and SOD) levels were measured by the ELISA method. The results showed that cerebral ischemia increased the apoptotic and necrotic cell death in the CA1 area of hippocampus, while, treatment with nesfatin-1significantly reduced apoptotic and necrotic cell death. Moreover, the MDA levels of the hippocampus in ischemic rats were higher, whereas in nesfatin-1-treated rats the MDA levels were decreased. Furthermore, the SOD and GSH levels in the ischemic rats were decreased, whilst in ischemic rats treated with nesfatin-1, the SOD and GSH levels were increased. This study for the first time found that nesfatin-1 treatment improves CA1 hippocampus injuries after cerebral ischemia through preventing neuronal cell death and enhancement of antioxidant defenses.
Collapse
Affiliation(s)
- Sohaila Erfani
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Ali Moghimi
- Rayan Center for Neuroscience and Behavior, Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Nahid Aboutaleb
- Physiology Research Center, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran.
- Department of Physiology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Mehdi Khaksari
- Addiction Research Center, Shahroud University of Medical Sciences, Shahroud, Iran
| |
Collapse
|
48
|
Roque C, Mendes-Oliveira J, Baltazar G. G protein-coupled estrogen receptor activates cell type-specific signaling pathways in cortical cultures: relevance to the selective loss of astrocytes. J Neurochem 2019; 149:27-40. [PMID: 30570746 DOI: 10.1111/jnc.14648] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 11/23/2018] [Accepted: 12/11/2018] [Indexed: 12/11/2022]
Abstract
Selective activation of the G protein-coupled estrogen receptor has been proposed to avoid some of the side effects elicited by the activation of classical estrogen receptors α and β. Although its contribution to neuroprotection triggered by estradiol in brain disorders has been explored, the results regarding ischemic stroke are contradictory, and currently, there is no consensus on the role that this receptor may play. The present study aimed to investigate the role of GPER in the ischemic insult. For that, primary cortical cultures exposed to oxygen and glucose deprivation (OGD) were used as a model. Our results demonstrate that neuronal survival was strongly affected by the ischemic insult and concurrent GPER activation with G1 had no further impact. In contrast, OGD had a smaller impact on astrocytes survival but G1, alone or combined with OGD, promoted their apoptosis. This effect was prevented by the GPER antagonist G15. The results also show that ischemia did not change the expression levels of GPER in neurons and astrocytes. In this study, we also demonstrate that selective activation of GPER induced astrocyte apoptosis via the phospholipase C pathway and subsequent intracellular calcium rise, whereas in neurons, this effect was not observed. Taken together, this evidence supports a direct impact of GPER activity on the viability of astrocytes, which seems to be associated with the regulation of different signaling pathways in astrocytes and neurons.
Collapse
Affiliation(s)
- Cláudio Roque
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | | | - Graça Baltazar
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal.,Faculty of Health Sciences, University of Beira Interior, Covilhã, Portugal
| |
Collapse
|
49
|
Butler MJ, Eckel LA. Eating as a motivated behavior: modulatory effect of high fat diets on energy homeostasis, reward processing and neuroinflammation. Integr Zool 2019; 13:673-686. [PMID: 29851251 DOI: 10.1111/1749-4877.12340] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Eating is a basic motivated behavior that provides fuel for the body and supports brain function. To ensure survival, the brain's feeding circuits are tuned to monitor peripheral energy balance and promote food-seeking behavior when energy stores are low. The brain's bias toward a positive energy state, which is necessary to ensure adequate nutrition during times of food scarcity, is evolutionarily conserved across mammalian species and is likely to drive overeating in the presence of a palatable, energy-dense diet. Animal models of diet-induced overeating have played a vital role in investigating how the drive to consume palatable food may override the homeostatic processes that serve to maintain energy balance. These animal models have provided valuable insights into the neurobiological mechanisms underlying homeostatic and non-homeostatic eating, motivation and food reward, and the development of obesity and related comorbidities. Here, we provide a brief review of this literature and discuss how diet-induced inflammation in the central nervous system impacts the neural control of food intake and regulation of body weight. The connection between diet and the immune system provides an exciting new direction for the study of ingestive behavior and the pathophysiology of obesity.
Collapse
Affiliation(s)
- Michael J Butler
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, Florida, USA
| | - Lisa A Eckel
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, Florida, USA
| |
Collapse
|
50
|
Hutson DD, Gurrala R, Ogola BO, Zimmerman MA, Mostany R, Satou R, Lindsey SH. Estrogen receptor profiles across tissues from male and female Rattus norvegicus. Biol Sex Differ 2019; 10:4. [PMID: 30635056 PMCID: PMC6329134 DOI: 10.1186/s13293-019-0219-9] [Citation(s) in RCA: 95] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 01/01/2019] [Indexed: 01/11/2023] Open
Abstract
Background Estrogen is formed by the enzyme aromatase (CYP19A1) and signals via three identified receptors ERα (ESR1), ERß (ESR2), and the G protein-coupled estrogen receptor (GPER). Understanding the relative contribution of each receptor to estrogenic signaling may elucidate the disparate effects of this sex hormone across tissues, and recent developments in PCR technology allow absolute quantification and direct comparison of multiple targets. We hypothesized that this approach would reveal tissue- and sex-specific differences in estrogen receptor mRNA. Methods ESR1, ESR2, GPER, and CYP19A1 were measured in four cardiovascular tissues (heart, aorta, kidney, and adrenal gland), three brain areas (somatosensory cortex, hippocampus, and prefrontal cortex), and reproductive tissues (ovaries, mammary gland, uterus, testes) from six male and six female adult Sprague-Dawley rats. Results GPER mRNA expression was relatively stable across all tissues in both sexes, ranging from 5.49 to 113 copies/ng RNA, a 21-fold difference. In contrast, ESR1/ESR2 were variable across tissues although similar within an organ system. ESR1 ranged from 4.46 to 614 copies/ng RNA (138-fold difference) while ESR2 ranged from 0.154 to 83.1 copies/ng RNA (540-fold). Significant sex differences were broadly absent except for renal ESR1 (female 206 vs. male 614 copies/ng RNA, P < 0.0001) and GPER (62.0 vs. 30.2 copies/ng RNA, P < 0.05) as well as gonadal GPER (5.49 vs. 47.5 copies/ng RNA, P < 0.01), ESR2 (83.1 vs. 0.299 copies/ng RNA, P < 0.01), and CYP19A1 (322 vs. 7.18 copies/ng RNA, P < 0.01). Cardiovascular tissues showed a predominance of ESR1, followed by GPER. In contrast, GPER was the predominant transcript in the brain with similarly low levels of ESR1 and ESR2. CYP19A1 was detected at very low levels except for reproductive tissues and the hippocampus. Conclusion While the data indicates a lack of sex differences in most tissues, significant differences were found in the range of receptor gene expression across tissues as well as in the receptor profile between organ systems. The data provide a guide for future studies by establishing estrogen receptor expression across multiple tissues using absolute PCR quantification. This knowledge on tissue-specific estrogen receptor profiles will aid the development of hormonal therapies that elicit beneficial effects in specific tissues.
Collapse
Affiliation(s)
- Dillion D Hutson
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Rakesh Gurrala
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Benard O Ogola
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Margaret A Zimmerman
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Ricardo Mostany
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, 70112, USA.,Tulane Brain Institute, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Ryousuke Satou
- Department of Physiology, Tulane University School of Medicine, New Orleans, LA, 70112, USA.,Hypertension and Renal Center of Excellence, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Sarah H Lindsey
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, 70112, USA. .,Tulane Brain Institute, Tulane University School of Medicine, New Orleans, LA, 70112, USA. .,Department of Physiology, Tulane University School of Medicine, New Orleans, LA, 70112, USA. .,Hypertension and Renal Center of Excellence, Tulane University School of Medicine, New Orleans, LA, 70112, USA.
| |
Collapse
|