1
|
Singh M, Krishnamoorthy VR, Kim S, Khurana S, LaPorte HM. Brain-derived neuerotrophic factor and related mechanisms that mediate and influence progesterone-induced neuroprotection. Front Endocrinol (Lausanne) 2024; 15:1286066. [PMID: 38469139 PMCID: PMC10925611 DOI: 10.3389/fendo.2024.1286066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 01/15/2024] [Indexed: 03/13/2024] Open
Abstract
Historically, progesterone has been studied significantly within the context of reproductive biology. However, there is now an abundance of evidence for its role in regions of the central nervous system (CNS) associated with such non-reproductive functions that include cognition and affect. Here, we describe mechanisms of progesterone action that support its brain-protective effects, and focus particularly on the role of neurotrophins (such as brain-derived neurotrophic factor, BDNF), the receptors that are critical for their regulation, and the role of certain microRNA in influencing the brain-protective effects of progesterone. In addition, we describe evidence to support the particular importance of glia in mediating the neuroprotective effects of progesterone. Through this review of these mechanisms and our own prior published work, we offer insight into why the effects of a progestin on brain protection may be dependent on the type of progestin (e.g., progesterone versus the synthetic, medroxyprogesterone acetate) used, and age, and as such, we offer insight into the future clinical implication of progesterone treatment for such disorders that include Alzheimer's disease, stroke, and traumatic brain injury.
Collapse
Affiliation(s)
- Meharvan Singh
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, United States
| | | | | | | | | |
Collapse
|
2
|
Jo SL, Hong EJ. Progesterone Receptor Membrane Component 1 Regulates Cellular Stress Responses and Inflammatory Pathways in Chronic Neuroinflammatory Conditions. Antioxidants (Basel) 2024; 13:230. [PMID: 38397828 PMCID: PMC10886208 DOI: 10.3390/antiox13020230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 02/08/2024] [Accepted: 02/11/2024] [Indexed: 02/25/2024] Open
Abstract
Alzheimer's disease (AD) is the leading cause of dementia and is one of the neurodegenerative diseases that are caused by neuronal death due to various triggers. Neuroinflammation plays a critical role in the development of AD. The neuroinflammatory response is manifested by pro-inflammatory cytokines, such as interleukin (IL)-1β, IL-6, and tumor necrosis factor-α; various chemokines; nitrous oxide; and reactive oxygen species. In this study, we evaluated the relevance of progesterone receptor membrane component 1 (PGRMC1), which is expressed in the brain cells during the induction of neuroinflammation. A lipopolysaccharide (LPS)-induced chronic neuroinflammation model and Pgrmc1 knockdown cells were used to assess the inflammatory cytokine levels, AD-related factors, inflammation-related signaling, and cell death. Pgrmc1 knockout (KO) mice had higher IL-1β levels after treatment with LPS compared with those of wild-type (WT) mice. Furthermore, Pgrmc1 KO mice had higher levels of inflammatory factors, endoplasmic reticulum stress indicators, and AD-associated markers compared with those of WT mice who underwent LPS treatment or not. Finally, these indicators were observed in vitro using U373-MG astrocytes. In conclusion, the loss of PGRMC1 may promote neuroinflammation and lead to AD.
Collapse
Affiliation(s)
| | - Eui-Ju Hong
- College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Republic of Korea;
| |
Collapse
|
3
|
Reddy DS, Singh T, Ramakrishnan S, Huber M, Wu X. Neuroprotectant Activity of Novel Water-Soluble Synthetic Neurosteroids on Organophosphate Intoxication and Status Epilepticus-Induced Long-Term Neurological Dysfunction, Neurodegeneration, and Neuroinflammation. J Pharmacol Exp Ther 2024; 388:399-415. [PMID: 38071567 PMCID: PMC10801736 DOI: 10.1124/jpet.123.001819] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 09/18/2023] [Indexed: 01/19/2024] Open
Abstract
Organophosphates (OPs) and nerve agents are potent neurotoxic compounds that cause seizures, status epilepticus (SE), brain injury, or death. There are persistent long-term neurologic and neurodegenerative effects that manifest months to years after the initial exposure. Current antidotes are ineffective in preventing these long-term neurobehavioral and neuropathological changes. Additionally, there are few effective neuroprotectants for mitigating the long-term effects of acute OP intoxication. We have pioneered neurosteroids as novel anticonvulsants and neuroprotectants for OP intoxication and seizures. In this study, we evaluated the efficacy of two novel synthetic, water-soluble neurosteroids, valaxanolone (VX) and lysaxanolone (LX), in combating the long-term behavioral and neuropathological impairments caused by acute OP intoxication and SE. Animals were exposed to the OP nerve agent surrogate diisopropylfluorophosphate (DFP) and were treated with VX or LX in addition to midazolam at 40 minutes postexposure. The extent of neurodegeneration, along with various behavioral and memory deficits, were assessed at 3 months postexposure. VX significantly reduced deficits of aggressive behavior, anxiety, memory, and depressive-like traits in control (DFP-exposed, midazolam-treated) animals; VX also significantly prevented the DFP-induced chronic loss of NeuN(+) principal neurons and PV(+) inhibitory neurons in the hippocampus and other regions. Additionally, VX-treated animals exhibited a reduced inflammatory response with decreased GFAP(+) astrogliosis and IBA1(+) microgliosis in the hippocampus, amygdala, and other regions. Similarly, LX showed significant improvement in behavioral and memory deficits, and reduced neurodegeneration and cellular neuroinflammation. Together, these results demonstrate the neuroprotectant effects of the novel synthetic neurosteroids in mitigating the long-term neurologic dysfunction and neurodegeneration associated with OP exposure. SIGNIFICANCE STATEMENT: Survivors of nerve agents and organophosphate (OP) exposures suffer from long-term neurological deficits. Currently, there is no specific drug therapy for mitigating the impact of OP exposure. However, novel synthetic neurosteroids that activate tonic inhibition provide a viable option for treating OP intoxication. The data from this study indicates the neuroprotective effects of synthetic, water-soluble neurosteroids for attenuation of long-term neurological deficits after OP intoxication. These findings establish valaxanolone and lysaxanolone as potent and efficacious neuroprotectants suitable for injectable dosing.
Collapse
Affiliation(s)
- Doodipala Samba Reddy
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University School of Medicine, Bryan, Texas (D.S.R., T.S., S.R., M.H., X.W.) and Institute of Pharmacology and Neurotherapeutics, Texas A&M University Health Science Center, Bryan, Texas (D.S.R., S.R., X.W.)
| | - Tanveer Singh
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University School of Medicine, Bryan, Texas (D.S.R., T.S., S.R., M.H., X.W.) and Institute of Pharmacology and Neurotherapeutics, Texas A&M University Health Science Center, Bryan, Texas (D.S.R., S.R., X.W.)
| | - Sreevidhya Ramakrishnan
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University School of Medicine, Bryan, Texas (D.S.R., T.S., S.R., M.H., X.W.) and Institute of Pharmacology and Neurotherapeutics, Texas A&M University Health Science Center, Bryan, Texas (D.S.R., S.R., X.W.)
| | - Madeline Huber
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University School of Medicine, Bryan, Texas (D.S.R., T.S., S.R., M.H., X.W.) and Institute of Pharmacology and Neurotherapeutics, Texas A&M University Health Science Center, Bryan, Texas (D.S.R., S.R., X.W.)
| | - Xin Wu
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University School of Medicine, Bryan, Texas (D.S.R., T.S., S.R., M.H., X.W.) and Institute of Pharmacology and Neurotherapeutics, Texas A&M University Health Science Center, Bryan, Texas (D.S.R., S.R., X.W.)
| |
Collapse
|
4
|
Kim S, Kubelka NK, LaPorte HM, Krishnamoorthy VR, Singh M. Estradiol and 3β-diol protect female cortical astrocytes by regulating connexin 43 Gap Junctions. Mol Cell Endocrinol 2023; 578:112045. [PMID: 37595662 PMCID: PMC10592012 DOI: 10.1016/j.mce.2023.112045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 08/03/2023] [Accepted: 08/14/2023] [Indexed: 08/20/2023]
Abstract
While estrogens have been described to protect or preserve neuronal function in the face of insults such as oxidative stress, the prevailing mechanistic model would suggest that these steroids exert direct effects on the neurons. However, there is growing evidence that glial cells, such as astrocytes, are key cellular mediators of protection. Noting that connexin 43 (Cx43), a protein highly expressed in astrocytes, plays a key role in mediating inter-cellular communication, we hypothesized that Cx43 is a target of estradiol (E2), and the estrogenic metabolite of DHT, 3β-diol. Additionally, we sought to determine if either or both of these hormones attenuate oxidative stress-induced cytotoxicity by eliciting a reduction in Cx43 expression or inhibition of Cx43 channel permeability. Using primary cortical astrocytes, we found that E2 and 3β-diol were each protective against the mixed metabolic/oxidative insult, iodoacetic acid (IAA). Moreover, these effects were blocked by estrogen receptor antagonists. However, E2 and 3β-diol did not alter Cx43 mRNA levels in astrocytes but did inhibit IAA-induced Cx43 gap junction opening/permeability. Taken together, these data implicate astrocyte Cx43 gap junction as an understudied mediator of the cytoprotective effects of estrogens in the brain. Given the wide breadth of disease states associated with Cx43 function/dysfunction, further understanding the relationship between gonadal steroids and Cx43 channels may contribute to a better understanding of the biological basis for sex differences in various diseases.
Collapse
Affiliation(s)
- Seongcheol Kim
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, United States
| | - Nicholas Knesek Kubelka
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, United States
| | - Heather M LaPorte
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, United States
| | - Vignesh R Krishnamoorthy
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, United States
| | - Meharvan Singh
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, United States.
| |
Collapse
|
5
|
Cao T, Wang L, Jiao S, Chen H, Lin C, Zhang B, Cai H. The Involvement of PGRMC1 Signaling in Cognitive Impairment Induced by Long-Term Clozapine Treatment in Rats. Neuropsychobiology 2023; 82:346-358. [PMID: 37673050 DOI: 10.1159/000533148] [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: 08/01/2022] [Accepted: 07/09/2023] [Indexed: 09/08/2023]
Abstract
INTRODUCTION Progesterone receptor component 1 (PGRMC1) has been identified as a potential target in atypical antipsychotic drug-induced metabolic disturbances as well as neuroprotection in the central nervous system. In our study, we aimed to figure out the essential role of PGRMC1 signaling pathway underlying clozapine-induced cognitive impairment. METHODS In male SD rats, we utilized recombinant adeno-associated viruses (BBB 2.0) and the specific inhibitor of PGRMC1 (AG205) to regulate the expression of PGRMC1 in the brain, with a special focus on the hippocampus. Treatments of clozapine and AG205 were conducted for 28 days, and subsequent behavioral tests including modified elevated plus maze and Morris water maze were conducted to evaluate the cognitive performance. Hippocampal protein expressions were measured by Western blotting. RESULTS Our study showed that long-term clozapine administration led to cognitive impairment as confirmed by behavioral tests as well as histopathological examination in the hippocampus. Clozapine inhibited neural survival through the PGRMC1/EGFR/GLP1R-PI3K-Akt signaling pathway, leading to a decrease in the downstream survival factor, brain-derived neurotrophic factor (BDNF), and simultaneously promoted neural apoptosis in the rat hippocampus. Intriguingly, by targeting at the hippocampal PGRMC1, we found that inhibiting PGRMC1 mimics, while its upregulation notably mitigates clozapine-induced cognitive impairment through PGRMC1 and its downstream signaling pathways. CONCLUSION PGRMC1-overexpression could protect hippocampus-dependent cognitive impairment induced by clozapine. This effect appears to arise, in part, from the upregulated expression of PGRMC1/EGFR/GLP1R and the activation of downstream PI3K-Akt-BDNF and caspase-3 signaling pathways.
Collapse
Affiliation(s)
- Ting Cao
- Department of Pharmacy, Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Second Xiangya Hospital, Central South University, Changsha, China
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - LiWei Wang
- Department of Pharmacy, Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Second Xiangya Hospital, Central South University, Changsha, China
| | - ShiMeng Jiao
- Department of Pharmacy, Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Second Xiangya Hospital, Central South University, Changsha, China
| | - Hui Chen
- Department of Pharmacy, Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Second Xiangya Hospital, Central South University, Changsha, China
| | - ChenQuan Lin
- Department of Pharmacy, Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Second Xiangya Hospital, Central South University, Changsha, China
| | - BiKui Zhang
- Department of Pharmacy, Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Second Xiangya Hospital, Central South University, Changsha, China
- International Research Center for Precision Medicine, Transformative Technology and Software Services, Hunan, China
| | - HuaLin Cai
- Department of Pharmacy, Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Second Xiangya Hospital, Central South University, Changsha, China
- International Research Center for Precision Medicine, Transformative Technology and Software Services, Hunan, China
| |
Collapse
|
6
|
Zhou C, Zhu T, Ni W, Zhou H, Song J, Wang M, Jin G, Zhou Y, Han J, Hua F. Gain-of-function of progesterone receptor membrane component 2 ameliorates ischemic brain injury. CNS Neurosci Ther 2023; 29:1585-1601. [PMID: 36794556 PMCID: PMC10173723 DOI: 10.1111/cns.14122] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/30/2023] [Accepted: 02/03/2023] [Indexed: 02/17/2023] Open
Abstract
OBJECTIVE Progesterone receptor membrane component 2 (PGRMC2) belongs to the membrane-associated progesterone receptor family, which regulates multiple pathophysiological processes. However, the role of PGRMC2 in ischemic stroke remains unexplored. The present study sought to determine the regulatory role of PGRMC2 in ischemic stroke. METHODS Male C57BL/6J mice were subjected to middle cerebral artery occlusion (MCAO). The protein expression level and localization of PGRMC2 were examined by western blotting and immunofluorescence staining. The gain-of-function ligand of PGRMC2 (CPAG-1, 45 mg/kg) was intraperitoneally injected into sham/MCAO mice, and brain infarction, blood-brain barrier (BBB) leakage, and sensorimotor functions were evaluated by magnetic resonance imaging, brain water content, Evans blue extravasation, immunofluorescence staining, and neurobehavioral tests. The astrocyte and microglial activation, neuronal functions, and gene expression profiles were revealed by RNA sequencing, qPCR, western blotting, and immunofluorescence staining after surgery and CPAG-1 treatment. RESULTS Progesterone receptor membrane component 2 was elevated in different brain cells after ischemic stroke. Intraperitoneal delivery of CPAG-1 reduced infarct size, brain edema, BBB leakage, astrocyte and microglial activation, and neuronal death, and improved sensorimotor deficits after ischemic stroke. CONCLUSION CPAG-1 acts as a novel neuroprotective compound that could reduce neuropathologic damage and improve functional recovery after ischemic stroke.
Collapse
Affiliation(s)
- Chao Zhou
- Institute of Neurological DiseasesXuzhou Medical UniversityXuzhouChina
- Department of NeurologyThe Affiliated Hospital of Xuzhou Medical UniversityXuzhouChina
| | - Taiyang Zhu
- Institute of Neurological DiseasesXuzhou Medical UniversityXuzhouChina
- Department of NeurologyThe Affiliated Hospital of Xuzhou Medical UniversityXuzhouChina
| | - Wanyan Ni
- Institute of Neurological DiseasesXuzhou Medical UniversityXuzhouChina
- Department of NeurologyThe Affiliated Hospital of Xuzhou Medical UniversityXuzhouChina
| | - Hui Zhou
- Institute of Neurological DiseasesXuzhou Medical UniversityXuzhouChina
- Department of NeurologyThe Affiliated Hospital of Xuzhou Medical UniversityXuzhouChina
| | - Jiaxing Song
- Department of NeurologyXinqiao Hospital and The Second Affiliated Hospital, Third Military Medical UniversityChongqingChina
| | - Miao Wang
- Department of GeriatricsThe Affiliated Hospital of Xuzhou Medical UniversityXuzhouChina
| | - Guoliang Jin
- Institute of Neurological DiseasesXuzhou Medical UniversityXuzhouChina
- Department of NeurologyThe Affiliated Hospital of Xuzhou Medical UniversityXuzhouChina
| | - Yan Zhou
- Institute of Neurological DiseasesXuzhou Medical UniversityXuzhouChina
- Department of NeurologyThe Affiliated Hospital of Xuzhou Medical UniversityXuzhouChina
| | - Jingjing Han
- Institute of Neurological DiseasesXuzhou Medical UniversityXuzhouChina
- Department of NeurologyThe Affiliated Hospital of Xuzhou Medical UniversityXuzhouChina
| | - Fang Hua
- Institute of Neurological DiseasesXuzhou Medical UniversityXuzhouChina
- Department of NeurologyThe Affiliated Hospital of Xuzhou Medical UniversityXuzhouChina
- Department of Interdisciplinary Health ScienceCollege of Allied Health Science, Augusta UniversityAugustaGeorgiaUSA
| |
Collapse
|
7
|
Pletzer B, Winkler-Crepaz K, Hillerer K. Progesterone and contraceptive progestin actions on the brain: A systematic review of animal studies and comparison to human neuroimaging studies. Front Neuroendocrinol 2023; 69:101060. [PMID: 36758768 DOI: 10.1016/j.yfrne.2023.101060] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 01/25/2023] [Accepted: 02/03/2023] [Indexed: 02/09/2023]
Abstract
In this review we systematically summarize the effects of progesterone and synthetic progestins on neurogenesis, synaptogenesis, myelination and six neurotransmitter systems. Several parallels between progesterone and older generation progestin actions emerged, suggesting actions via progesterone receptors. However, existing results suggest a general lack of knowledge regarding the effects of currently used progestins in hormonal contraception regarding these cellular and molecular brain parameters. Human neuroimaging studies were reviewed with a focus on randomized placebo-controlled trials and cross-sectional studies controlling for progestin type. The prefrontal cortex, amygdala, salience network and hippocampus were identified as regions of interest for future preclinical studies. This review proposes a series of experiments to elucidate the cellular and molecular actions of contraceptive progestins in these areas and link these actions to behavioral markers of emotional and cognitive functioning. Emotional effects of contraceptive progestins appear to be related to 1) alterations in the serotonergic system, 2) direct/indirect modulations of inhibitory GABA-ergic signalling via effects on the allopregnanolone content of the brain, which differ between androgenic and anti-androgenic progestins. Cognitive effects of combined oral contraceptives appear to depend on the ethinylestradiol dose.
Collapse
Affiliation(s)
- Belinda Pletzer
- Department of Psychology & Centre for Cognitive Neuroscience, Paris-Lodron-University Salzburg, Salzburg Austria.
| | | | - Katharina Hillerer
- Department of Gynaecology & Obstetrics, Private Medical University, Salzburg, Austria
| |
Collapse
|
8
|
Involvement of DAAO Overexpression in Delayed Hippocampal Neuronal Death. Cells 2022; 11:cells11223689. [PMID: 36429117 PMCID: PMC9688509 DOI: 10.3390/cells11223689] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 11/17/2022] [Accepted: 11/17/2022] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND D-amino acid oxidase (DAAO) is a flavoenzyme that specifically catalyzes the deamination of many neutral and basic D-amino acids. This study aims to explore the pathological increment of hippocampal DAAO and its potential relationship with delayed hippocampal neuronal death. METHODS Ischemia-reperfusion was induced in mice through middle cerebral artery occlusion (MCAO). Neurological deficit scores and hippocampal neuronal death were assessed in MCAO mice. Immunofluorescent staining was applied to identify activated astrocytes and evaluate DAAO expression. TUNEL and Nissl staining were utilized to identify cell apoptosis of hippocampal neurons. RESULTS Hippocampal astrocytic DAAO was strikingly increased following ischemic stroke, with the greatest increase on day 5 after surgery, followed by the manifestation of neurobehavioral deficits. Astrocytic DAAO was found to be mainly expressed in the hippocampal CA2 region and linked with subsequent specific neural apoptosis. Thus, it is supposed that the activation of astrocytic DAAO in ischemic stroke might contribute to neuronal death. An intravenous, twice-daily administration of 4H-furo[3,2-b]pyrrole-5-carboxylic acid (SUN, 10 mg/kg) markedly relieved behavioral status and delayed hippocampal neuronal death by 38.0% and 41.5%, respectively, compared to the model group treated with saline. In transfected primary astrocytes, DAAO overexpression inhibits cell activity, induces cytotoxicity, and promotes hippocampal neuronal death at least partly by enhancing H2O2 levels with subsequent activation of TRP calcium channels in neurons. CONCLUSIONS Our findings suggest that increased hippocampal DAAO is causally associated with the development of delayed neuronal death after MCAO onset via astrocyte-neuron interactions. Hence, targeting DAAO is a promising therapeutic strategy for the management of neurological disorders.
Collapse
|
9
|
Murray TE, Richards CM, Robert-Gostlin VN, Bernath AK, Lindhout IA, Klegeris A. Potential neurotoxic activity of diverse molecules released by astrocytes. Brain Res Bull 2022; 189:80-101. [PMID: 35988785 DOI: 10.1016/j.brainresbull.2022.08.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 07/04/2022] [Accepted: 08/14/2022] [Indexed: 11/02/2022]
Abstract
Astrocytes are the main support cells of the central nervous system. They also participate in neuroimmune reactions. In response to pathological and immune stimuli, astrocytes transform to reactive states characterized by increased release of inflammatory mediators. Some of these molecules are neuroprotective and inflammation resolving while others, including reactive oxygen species (ROS), nitric oxide (NO), matrix metalloproteinase (MMP)- 9, L-glutamate, and tumor necrosis factor α (TNF), are well-established toxins known to cause damage to surrounding cells and tissues. We hypothesized that similar to microglia, the brain immune cells, reactive astrocytes can release a broader set of diverse molecules that are potentially neurotoxic. A literature search was conducted to identify such molecules using the following two criteria: 1) evidence of their expression and secretion by astrocytes and 2) direct neurotoxic action. This review describes 14 structurally diverse molecules as less-established astrocyte neurotoxins, including C-X-C motif chemokine ligand (CXCL)10, CXCL12/CXCL12(5-67), FS-7-associated surface antigen ligand (FasL), macrophage inflammatory protein (MIP)- 2α, TNF-related apoptosis inducing ligand (TRAIL), pro-nerve growth factor (proNGF), pro-brain-derived neurotrophic factor (proBDNF), chondroitin sulfate proteoglycans (CSPGs), cathepsin (Cat)B, group IIA secretory phospholipase A2 (sPLA2-IIA), amyloid beta peptides (Aβ), high mobility group box (HMGB)1, ceramides, and lipocalin (LCN)2. For some of these molecules, further studies are required to establish either their direct neurotoxic effects or the full spectrum of stimuli that induce their release by astrocytes. Only limited studies with human-derived astrocytes and neurons are available for most of these potential neurotoxins, which is a knowledge gap that should be addressed in the future. We also summarize available evidence of the role these molecules play in select neuropathologies where reactive astrocytes are a key feature. A comprehensive understanding of the full spectrum of neurotoxins released by reactive astrocytes is key to understanding neuroinflammatory diseases characterized by the adverse activation of these cells and may guide the development of novel treatment strategies.
Collapse
Affiliation(s)
- Taryn E Murray
- Department of Biology, University of British Columbia Okanagan Campus, Kelowna, British Columbia V1V 1V7, Canada
| | - Christy M Richards
- Department of Biology, University of British Columbia Okanagan Campus, Kelowna, British Columbia V1V 1V7, Canada
| | - Victoria N Robert-Gostlin
- Department of Biology, University of British Columbia Okanagan Campus, Kelowna, British Columbia V1V 1V7, Canada
| | - Anna K Bernath
- Department of Biology, University of British Columbia Okanagan Campus, Kelowna, British Columbia V1V 1V7, Canada
| | - Ivan A Lindhout
- Department of Biology, University of British Columbia Okanagan Campus, Kelowna, British Columbia V1V 1V7, Canada
| | - Andis Klegeris
- Department of Biology, University of British Columbia Okanagan Campus, Kelowna, British Columbia V1V 1V7, Canada.
| |
Collapse
|
10
|
Sun X, Hu Y, Zhou H, Wang S, Zhou C, Lin L, Zhu T, Ge J, Han J, Zhou Y, Jin G, Wang Y, Zu J, Shi H, Yang X, Zan K, Wang J, Hua F. Inhibition of progesterone receptor membrane component-1 exacerbates neonatal hypoxic-ischemic cerebral damage in male mice. Exp Neurol 2021; 347:113893. [PMID: 34653511 DOI: 10.1016/j.expneurol.2021.113893] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 10/04/2021] [Accepted: 10/08/2021] [Indexed: 11/04/2022]
Abstract
This study investigated the expression of progesterone receptor membrane component 1 (pgrmc1) in the brains of male and female mice, and the effect of inhibiting pgrmc1 on neonatal hypoxic-ischemic (HI) cerebral injury in male mice. A mouse model of neonatal HI brain injury was established, and AG205, a specific antagonist of pgrmc1, was injected into the left lateral cerebral ventricle 1 h before HI. Histological staining, behavior testing, Western blots, and quantitative PCR (qPCR) were employed to evaluate pgrmc1 expression, brain damage, neurological function, and molecular mechanisms. Results demonstrated that the mRNA and protein levels of pgrmc1 increased significantly in the cortex and hippocampus 72 h after HI without sex differences. The inhibition of pgrmc1 exacerbated the neonatal brain damage in the acute stage of HI in male mice as seen in the increase in brain water content, infarction area, and neuronal death. Inhibition of pgrmc1 also aggravated the neurological dysfunction and anxiety induced by HI brain injury. In addition, inhibition of pgrmc1 activated the NF-kB signaling and NF-κB-mediated cytokines, and inhibited BDNF/PI3K/AKT pathway in the brains of the newborn HI mice. The results indicated that pgrmc1 inhibition exacerbated the brain damage in newborn male mice subjected to HI by activating IκBα/NFκB signaling and inhibiting BDNF/PI3K/Akt pathway.
Collapse
Affiliation(s)
- Xiaoyu Sun
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Department of Rehabilitation Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yuting Hu
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Department of Rehabilitation Medicine, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Hui Zhou
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Institute of Neurological Diseases, Xuzhou Medical University, Xuzhou, China
| | - Shang Wang
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Institute of Neurological Diseases, Xuzhou Medical University, Xuzhou, China
| | - Chao Zhou
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Institute of Neurological Diseases, Xuzhou Medical University, Xuzhou, China
| | - Li Lin
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Institute of Neurological Diseases, Xuzhou Medical University, Xuzhou, China
| | - Taiyang Zhu
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Institute of Neurological Diseases, Xuzhou Medical University, Xuzhou, China
| | - Ji Ge
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Institute of Neurological Diseases, Xuzhou Medical University, Xuzhou, China
| | - Jingjing Han
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Institute of Neurological Diseases, Xuzhou Medical University, Xuzhou, China
| | - Yan Zhou
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Institute of Neurological Diseases, Xuzhou Medical University, Xuzhou, China
| | - Guoliang Jin
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Institute of Neurological Diseases, Xuzhou Medical University, Xuzhou, China
| | - Yuqiao Wang
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Institute of Neurological Diseases, Xuzhou Medical University, Xuzhou, China
| | - Jie Zu
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Institute of Neurological Diseases, Xuzhou Medical University, Xuzhou, China
| | - Hongjuan Shi
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Institute of Neurological Diseases, Xuzhou Medical University, Xuzhou, China
| | - Xingxing Yang
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Institute of Neurological Diseases, Xuzhou Medical University, Xuzhou, China
| | - Kun Zan
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Institute of Neurological Diseases, Xuzhou Medical University, Xuzhou, China
| | - Jun Wang
- Department of Pediatrics, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Fang Hua
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Institute of Neurological Diseases, Xuzhou Medical University, Xuzhou, China.
| |
Collapse
|
11
|
Castelnovo LF, Thomas P. Membrane progesterone receptor α (mPRα/PAQR7) promotes migration, proliferation and BDNF release in human Schwann cell-like differentiated adipose stem cells. Mol Cell Endocrinol 2021; 531:111298. [PMID: 33930460 DOI: 10.1016/j.mce.2021.111298] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 04/20/2021] [Accepted: 04/21/2021] [Indexed: 12/26/2022]
Abstract
Membrane progesterone receptors (mPRs) were recently found to be present and active in Schwann cells, where they have a potentially pro-regenerative activity. In this study, we investigated the role of mPRs in human adipose stem cells (ASC) differentiated into Schwann cell-like cells (SCL-ASC), which represent a promising alternative to Schwann cells for peripheral nerve regeneration. Our findings show that mPRs are present both in undifferentiated and differentiated ASC, and that the differentiation protocol upregulates mPR expression. Activation of mPRα promoted cell migration and differentiation in SCL-ASC, alongside with changes in cell morphology and mPRα localization. Moreover, it increased the expression and release of BDNF, a neurotrophin with pro-regenerative activity. Further analysis showed that Src and PI3K-Akt signaling pathways are involved in mPRα activity in SCL-ASC. These findings suggest that mPRα could play a pro-regenerative role in SCL-ASC and may be a promising target for the promotion of peripheral nerve regeneration.
Collapse
Affiliation(s)
- Luca F Castelnovo
- Marine Science Institute, The University of Texas at Austin, 750 Channel View Drive, Port Aransas (TX), 78373, United States.
| | - Peter Thomas
- Marine Science Institute, The University of Texas at Austin, 750 Channel View Drive, Port Aransas (TX), 78373, United States
| |
Collapse
|
12
|
Xu YH, Wang XX, Wang MJ, Liu YY, Xue Z, Chen JX. Influence of progestational stress on BDNF and NMDARs in the hippocampus of male offspring and amelioration by Chaihu Shugan San. Biomed Pharmacother 2021; 135:111204. [PMID: 33548869 DOI: 10.1016/j.biopha.2020.111204] [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: 08/11/2020] [Revised: 12/13/2020] [Accepted: 12/26/2020] [Indexed: 12/09/2022] Open
Abstract
BACKGROUND Progestational stress has been proven to be a risk for the neural development of offspring, especially in the hippocampus. However, whether Chaihu Shugan San (CSS) can ameliorate hippocampal neural development via the regulation of brain-derived neurotrophic factor (BDNF), and N-methyl-D-aspartate receptors (NMDAR) 2A (NR2A) and 2B (NR2B), and the mechanism of such action remains unclear. METHODS Thirty-six female rats were randomly allocated into control, chronic immobilization stress (CIS) and CSS groups according to the random number table, respectively. The male offspring were fed for 21 days after birth then randomly divided into the same three groups (6 rats/group) as the female rats. Female rats, except for the control group, underwent 21-day CIS to established a progestational stress anxiety-like model which was evaluated by body weight, the elevated plus-maze (EPM) test and serum dopamine (DA) measured using an enzyme-linked immunosorbent assay (ELISA). The expression levels of estrogen receptors (ERα/ERβ) and progesterone receptor (PR) in female rat ovaries were quantified by real-time fluorescence quantitative polymerase chain reaction (RT-qPCR) and Western blot analysis. The hippocampal tissue in the 21-day offspring was observed by hematoxylin-eosin (HE) staining. The concentration of BDNF, NR2A, and NR2B were measured by RT-qPCR and immunohistochemistry in the CA3 and dentate gyrus (DG) regions of offsprings' hippocampus. RESULTS Compared with the female control group, significant differences in body weight, EPM test and DA concentration were observed in the CIS group, meanwhile, the concentration of ERα (P < 0.05), PR (P < 0.05) and ERβ in the ovaries were decreased. In the offsprings' hippocampus of the CIS group, the chromatin of the nucleus was edge set and with condensed and irregular morphology nucleus, and the cytoplasm was unevenly stained with spaces around the cells, moreover, the expression levels of BDNF, NR2A, and NR2B were also declined (P < 0.05). However, Chaihu Shugan San reversed these changes, especially the BDNF in the DG region (P < 0.05), and NR2A and NR2B in the CA3 and DG region (P < 0.05). CONCLUSIONS CSS could ameliorate the neural development of the hippocampus in offspring damaged by anxiety-like progestational stress in female rats via regulating the expression levels of ERα, ERβ, and PR in female rat ovaries and BDNF, NR2A, and NR2B in the hippocampus of their offspring.
Collapse
MESH Headings
- Animals
- Brain-Derived Neurotrophic Factor/genetics
- Brain-Derived Neurotrophic Factor/metabolism
- Disease Models, Animal
- Estrogen Receptor alpha/genetics
- Estrogen Receptor alpha/metabolism
- Estrogen Receptor beta/genetics
- Estrogen Receptor beta/metabolism
- Female
- Gestational Age
- Hippocampus/drug effects
- Hippocampus/metabolism
- Hippocampus/pathology
- Male
- Neurogenesis/drug effects
- Ovary/drug effects
- Ovary/metabolism
- Plant Extracts/pharmacology
- Pregnancy
- Prenatal Exposure Delayed Effects
- Rats, Wistar
- Receptors, N-Methyl-D-Aspartate/genetics
- Receptors, N-Methyl-D-Aspartate/metabolism
- Receptors, Progesterone/genetics
- Receptors, Progesterone/metabolism
- Restraint, Physical
- Signal Transduction
- Stress, Psychological/drug therapy
- Stress, Psychological/genetics
- Stress, Psychological/metabolism
- Stress, Psychological/pathology
- Rats
Collapse
Affiliation(s)
- Ya-Hui Xu
- School of Basic Medicine, Henan University of Chinese Medicine, Zhengzhou, 450046, Henan, China; School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Xin-Xing Wang
- School of Basic Medicine, Henan University of Chinese Medicine, Zhengzhou, 450046, Henan, China
| | - Ming-Jing Wang
- School of Basic Medicine, Henan University of Chinese Medicine, Zhengzhou, 450046, Henan, China
| | - Yue-Yun Liu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Zhe Xue
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Jia-Xu Chen
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China; Formula-Pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632, Guangdong, China.
| |
Collapse
|
13
|
Rurak GM, Woodside B, Aguilar-Valles A, Salmaso N. Astroglial cells as neuroendocrine targets in forebrain development: Implications for sex differences in psychiatric disease. Front Neuroendocrinol 2021; 60:100897. [PMID: 33359797 DOI: 10.1016/j.yfrne.2020.100897] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/05/2020] [Accepted: 12/15/2020] [Indexed: 12/23/2022]
Abstract
Astroglial cells are the most abundant cell type in the mammalian brain. They are implicated in almost every aspect of brain physiology, including maintaining homeostasis, building and maintaining the blood brain barrier, and the development and maturation of neuronal networks. Critically, astroglia also express receptors for gonadal sex hormones, respond rapidly to gonadal hormones, and are able to synthesize hormones. Thus, they are positioned to guide and mediate sexual differentiation of the brain, particularly neuronal networks in typical and pathological conditions. In this review, we describe astroglial involvement in the organization and development of the brain, and consider known sex differences in astroglial responses to understand how astroglial cell-mediated organization may play a role in forebrain sexual dimorphisms in human populations. Finally, we consider how sexually dimorphic astroglial responses and functions in development may lead to sex differences in vulnerability for neuropsychiatric disorders.
Collapse
Affiliation(s)
- Gareth M Rurak
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| | - Barbara Woodside
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada; Concordia University, Montreal, Quebec, Canada
| | | | - Natalina Salmaso
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada.
| |
Collapse
|
14
|
Resveratrol reserved hypoxia-ischemia induced childhood hippocampal dysfunction and neurogenesis via improving mitochondrial dynamics. Neurosci Res 2020; 161:51-58. [DOI: 10.1016/j.neures.2019.11.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 11/22/2019] [Accepted: 11/27/2019] [Indexed: 12/30/2022]
|
15
|
Abstract
The neurotrophic factor BDNF is an important regulator for the development of brain circuits, for synaptic and neuronal network plasticity, as well as for neuroregeneration and neuroprotection. Up- and downregulations of BDNF levels in human blood and tissue are associated with, e.g., neurodegenerative, neurological, or even cardiovascular diseases. The changes in BDNF concentration are caused by altered dynamics in BDNF expression and release. To understand the relevance of major variations of BDNF levels, detailed knowledge regarding physiological and pathophysiological stimuli affecting intra- and extracellular BDNF concentration is important. Most work addressing the molecular and cellular regulation of BDNF expression and release have been performed in neuronal preparations. Therefore, this review will summarize the stimuli inducing release of BDNF, as well as molecular mechanisms regulating the efficacy of BDNF release, with a focus on cells originating from the brain. Further, we will discuss the current knowledge about the distinct stimuli eliciting regulated release of BDNF under physiological conditions.
Collapse
Affiliation(s)
- Tanja Brigadski
- Department of Informatics and Microsystem Technology, University of Applied Sciences Kaiserslautern, D-66482, Zweibrücken, Germany.
| | - Volkmar Leßmann
- Institute of Physiology, Otto-von-Guericke University, D-39120, Magdeburg, Germany.
- Center for Behavioral Brain Sciences, Magdeburg, Germany.
| |
Collapse
|
16
|
Wu Z, Wu H, Sun S, Wu H, Shi W, Song J, Liu J, Zhang Y, Bian F, Jia P, Hou Y. Progesterone attenuates Aβ25–35-induced neuronal toxicity by activating the Ras signalling pathway through progesterone receptor membrane component 1. Life Sci 2020; 253:117360. [DOI: 10.1016/j.lfs.2020.117360] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 01/16/2020] [Accepted: 01/24/2020] [Indexed: 12/23/2022]
|
17
|
Gonzalez A. Antioxidants and Neuron-Astrocyte Interplay in Brain Physiology: Melatonin, a Neighbor to Rely on. Neurochem Res 2020; 46:34-50. [PMID: 31989469 DOI: 10.1007/s11064-020-02972-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 01/19/2020] [Accepted: 01/21/2020] [Indexed: 12/19/2022]
Abstract
This manuscript is a review focused onto the role of astrocytes in the protection of neurons against oxidative stress and how melatonin can contribute to the maintenance of brain homeostasis. The first part of the review is dedicated to the dependence of neurons on astrocytes by terms of survival under oxidative stress conditions. Additionally, the effects of melatonin against oxidative stress in the brain and its putative role in the protection against diseases affecting the brain are highlighted. The effects of melatonin on the physiology of neurons and astrocytes also are reviewed.
Collapse
Affiliation(s)
- Antonio Gonzalez
- Department of Physiology, Institute of Molecular Pathology Biomarkers, University of Extremadura, Avenida de las Ciencias s/n, 10003, Cáceres, Spain.
| |
Collapse
|
18
|
Yilmaz C, Karali K, Fodelianaki G, Gravanis A, Chavakis T, Charalampopoulos I, Alexaki VI. Neurosteroids as regulators of neuroinflammation. Front Neuroendocrinol 2019; 55:100788. [PMID: 31513776 DOI: 10.1016/j.yfrne.2019.100788] [Citation(s) in RCA: 128] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 08/12/2019] [Accepted: 09/07/2019] [Indexed: 02/07/2023]
Abstract
Neuroinflammation is a physiological protective response in the context of infection and injury. However, neuroinflammation, especially if chronic, may also drive neurodegeneration. Neurodegenerative diseases, such as multiple sclerosis (MS), Alzheimer's disease (AD), Parkinson's disease (PD) and traumatic brain injury (TBI), display inflammatory activation of microglia and astrocytes. Intriguingly, the central nervous system (CNS) is a highly steroidogenic environment synthesizing steroids de novo, as well as metabolizing steroids deriving from the circulation. Neurosteroid synthesis can be substantially affected by neuroinflammation, while, in turn, several steroids, such as 17β-estradiol, dehydroepiandrosterone (DHEA) and allopregnanolone, can regulate neuroinflammatory responses. Here, we review the role of neurosteroids in neuroinflammation in the context of MS, AD, PD and TBI and describe underlying molecular mechanisms. Moreover, we introduce the concept that synthetic neurosteroid analogues could be potentially utilized for the treatment of neurodegenerative diseases in the future.
Collapse
Affiliation(s)
- Canelif Yilmaz
- Institute of Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany
| | - Kanelina Karali
- Department of Pharmacology, Medical School, University of Crete, Heraklion, Greece; Institute of Molecular Biology & Biotechnology, Foundation of Research & Technology-Hellas, Heraklion, Greece
| | - Georgia Fodelianaki
- Institute of Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany
| | - Achille Gravanis
- Department of Pharmacology, Medical School, University of Crete, Heraklion, Greece; Institute of Molecular Biology & Biotechnology, Foundation of Research & Technology-Hellas, Heraklion, Greece
| | - Triantafyllos Chavakis
- Institute of Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany; Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Ioannis Charalampopoulos
- Department of Pharmacology, Medical School, University of Crete, Heraklion, Greece; Institute of Molecular Biology & Biotechnology, Foundation of Research & Technology-Hellas, Heraklion, Greece
| | - Vasileia Ismini Alexaki
- Institute of Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany.
| |
Collapse
|
19
|
Tanaka M, Ogaeri T, Samsonov M, Sokabe M. Nestorone exerts long-term neuroprotective effects against transient focal cerebral ischemia in adult male rats. Brain Res 2019; 1719:288-296. [DOI: 10.1016/j.brainres.2018.09.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 09/18/2018] [Accepted: 09/18/2018] [Indexed: 11/17/2022]
|
20
|
Yin H, Jiang Y, Zhang Y, Ge H, Yang Z. The inhibition of BDNF/TrkB/PI3K/Akt signal mediated by AG1601 promotes apoptosis in malignant glioma. J Cell Biochem 2019; 120:18771-18781. [PMID: 31219215 DOI: 10.1002/jcb.29190] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Accepted: 05/28/2019] [Indexed: 12/15/2022]
Abstract
Malignant glioma is the most aggressive primary brain tumor and has a poor survival rate. Even if extensive methods are preformed to treat glioma, the mortality rate is still very high. It is necessary for discovering and developing new drugs for malignant glioma treatment. AG1601 is one of AG-series drugs, including AG1031 and AG1503, and it has been optimized on the original basis. In our study, we found that AG1601 markedly inhibited proliferation and promoted C6 glioma cell apoptosis in vitro. AG1601 also reduced the size and weight of glioma in vivo. The growth ability of glioma was significantly inhibited after treatment with AG1601. It also showed that the expression levels of BDNF/TrkB/PI3K/Akt signal related proteins were obviously decreased in C6 glioma cells after treatment with AG1601 in vivo and in vitro. We also found that BDNF, as the activator of BDNF/TrkB/PI3K/Akt signal, reversed the anti-proliferation and pro-apoptosis of C6 glioma cells caused by AG1601. K252a, a specific inhibitor of TrkB, and AG1601 in combination aggravated C6 glioma cell apoptosis. These results indicate that AG1601 has good effects on the anti-proliferation and pro-apoptosis of malignant glioma via BDNF/TrkB/PI3K/Akt signal and could be considered as a potential drug in treating malignant glioma.
Collapse
Affiliation(s)
- Hongqiang Yin
- Medical School, Key Laboratory of Bioactive Materials for Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China
| | - Yu Jiang
- Medical School, Key Laboratory of Bioactive Materials for Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China
| | - Yinguo Zhang
- Department of Physiology and Pathophysiology, Logistics University of Chinese People's Armed Police Force, Tianjin, China
| | - Hui Ge
- Division of drug discovery, AscentGene Inc, Gaithersburg, Maryland
| | - Zhuo Yang
- Medical School, Key Laboratory of Bioactive Materials for Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China
| |
Collapse
|
21
|
Let-7i inhibition enhances progesterone-induced functional recovery in a mouse model of ischemia. Proc Natl Acad Sci U S A 2018; 115:E9668-E9677. [PMID: 30237284 DOI: 10.1073/pnas.1803384115] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Progesterone (P4) is a potent neuroprotectant and a promising therapeutic for stroke treatment. However, the underlying mechanism(s) remain unclear. Our laboratory recently reported that brain-derived neurotrophic factor (BDNF) is a critical mediator of P4's protective actions and that P4-induced BDNF release from cortical astrocytes is mediated by a membrane-associated progesterone receptor, Pgrmc1. Here, we report that the microRNA (miRNA) let-7i is a negative regulator of Pgrmc1 and BDNF in glia and that let-7i disrupts P4-induced BDNF release and P4's beneficial effects on cell viability and markers of synaptogenesis. Using an in vivo model of ischemia, we demonstrate that inhibiting let-7i enhances P4-induced neuroprotection and facilitates functional recovery following stroke. The discovery of such factors that regulate the cytoprotective effects of P4 may lead to the development of biomarkers to differentiate/predict those likely to respond favorably to P4 versus those that do not.
Collapse
|
22
|
Feng L, Allen TK, Marinello WP, Murtha AP. Roles of Progesterone Receptor Membrane Component 1 in Oxidative Stress-Induced Aging in Chorion Cells. Reprod Sci 2018; 26:394-403. [PMID: 29783884 DOI: 10.1177/1933719118776790] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
INTRODUCTION Oxidative stress-mediated fetal membrane cell aging is activated prematurely in preterm premature rupture of membranes (PPROMs). The mechanism of this phenomenon is largely understudied. Progesterone receptor membrane component 1 (PGRMC1) has been recognized as a potential protective component for maintaining fetal membrane integrity and healthy pregnancies. We aimed to investigate the effects of oxidative stress (represented by hydrogen peroxide [H2O2]) on fetal membrane and chorion cell senescence, p38 mitogen-activated protein kinase (MAPK) phosphorylation, and sirtuin 3 (SIRT3) and to examine the roles of PGRMC1 in these effects. METHODS Following serum starvation for 24 hours, full-thickness fetal membrane explants and primary chorion cells were treated with H2O2 at 100, 300, and 500 µM for 24 hours. Cells were fixed for cell senescence-associated β-galactosidase assay. Cell lysates were harvested for quantitive reverse transcription polymerase chain reaction to quantify SIRT3 messenger RNA. Cell lysates were harvested for Western blot to semi-quantify SIRT3 protein and p38 MAPK phosphorylation levels, respectively. To examine the role of PGRMC1, primary chorion cells underwent the same treatment mentioned above following PGRMC1 knockdown using validated PGRMC1-specific small-interfering RNA. RESULTS Hydrogen peroxide significantly induced cell senescence and p38 MAPK phosphorylation, and it significantly decreased SIRT3 expression in full-thickness fetal membrane explants and chorion cells. These effects were enhanced by PGRMC1 knockdown. DISCUSSION This study further demonstrated that oxidative stress-induced cell aging is one of the mechanisms of PPROM and PGRMC1 acts as a protective element for maintaining fetal membrane integrity by inhibiting oxidative stress-induced chorion cell aging.
Collapse
Affiliation(s)
- Liping Feng
- 1 Department of Obstetrics and Gynecology, Duke University, Durham, NC, USA.,2 Ministry of Education and Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Terrence K Allen
- 3 Department of Anesthesiology, Duke University, Durham, NC, USA
| | | | - Amy P Murtha
- 1 Department of Obstetrics and Gynecology, Duke University, Durham, NC, USA
| |
Collapse
|
23
|
Hampton KK, Anderson K, Frazier H, Thibault O, Craven RJ. Insulin Receptor Plasma Membrane Levels Increased by the Progesterone Receptor Membrane Component 1. Mol Pharmacol 2018; 94:665-673. [PMID: 29674524 DOI: 10.1124/mol.117.110510] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 04/13/2018] [Indexed: 12/14/2022] Open
Abstract
The insulin receptor (IR) is a ligand-activated receptor tyrosine kinase that has a key role in metabolism, cellular survival, and proliferation. Progesterone receptor membrane component 1 (PGRMC1) promotes cellular signaling via receptor trafficking and is essential for some elements of tumor growth and metastasis. In the present study, we demonstrate that PGRMC1 coprecipitates with IR. Furthermore, we show that PGRMC1 increases plasma membrane IR levels in multiple cell lines and decreases insulin binding at the cell surface. The findings have therapeutic applications because a small-molecule PGRMC1 ligand, AG205, also decreases plasma membrane IR levels. However, PGRMC1 knockdown via short hairpin RNA expression and AG205 treatment potentiated insulin-mediated phosphorylation of the IR signaling mediator AKT. Finally, PGRMC1 also increased plasma membrane levels of two key glucose transporters, GLUT-4 and GLUT-1. Our data support a role for PGRMC1 maintaining plasma membrane pools of the receptor, modulating IR signaling and function.
Collapse
Affiliation(s)
- Kaia K Hampton
- Department of Pharmacology and Nutritional Sciences, Markey Cancer Center, University of Kentucky College of Medicine, Lexington, Kentucky
| | - Katie Anderson
- Department of Pharmacology and Nutritional Sciences, Markey Cancer Center, University of Kentucky College of Medicine, Lexington, Kentucky
| | - Hilaree Frazier
- Department of Pharmacology and Nutritional Sciences, Markey Cancer Center, University of Kentucky College of Medicine, Lexington, Kentucky
| | - Olivier Thibault
- Department of Pharmacology and Nutritional Sciences, Markey Cancer Center, University of Kentucky College of Medicine, Lexington, Kentucky
| | - Rolf J Craven
- Department of Pharmacology and Nutritional Sciences, Markey Cancer Center, University of Kentucky College of Medicine, Lexington, Kentucky
| |
Collapse
|
24
|
Kabe Y, Handa H, Suematsu M. Function and structural regulation of the carbon monoxide (CO)-responsive membrane protein PGRMC1. J Clin Biochem Nutr 2018; 63:12-17. [PMID: 30087538 PMCID: PMC6064819 DOI: 10.3164/jcbn.17-132] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 01/04/2018] [Indexed: 01/29/2023] Open
Abstract
Progesterone receptor membrane associated component 1 is a multifunctional heme-binding protein that plays a role in several biological processes such as tumor progression, metabolic regulation, and viability control of nerve cells. Notably, progesterone receptor membrane associated component 1 is highly expressed in various types of cancer cells, and facilitates cancer proliferation and chemoresistance. Recently, progesterone receptor membrane associated component 1 structure has been explored by X-ray crystallographic analysis. Interestingly, whereas apo- progesterone receptor membrane associated component 1 exists as a monomer, the heme-bound progesterone receptor membrane associated component 1 converts into a stable dimer by forming a unique heme-heme stacking structure, leading to activation of epidermal growth factor receptor signaling and chemoresistance in cancer cells. Furthermore, the gas mediator carbon monoxide inhibits progesterone receptor membrane associated component 1-mediated activation in cancer cells by dissociating the heme-stacking dimer of progesterone receptor membrane associated component 1. The dynamic structural regulation of progesterone receptor membrane associated component 1 will provide new insights for understanding the mechanisms underlying its various functions.
Collapse
Affiliation(s)
- Yasuaki Kabe
- Department of Biochemistry, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan.,Core Research for Evolutional Science and Technology (CREST), Japan Agency for Medical Research and Development (AMED), 20F Yomiuri Shimbun Bldg, 1-7-1 Otemachi, Chiyoda-ku, Tokyo 100-0004, Japan
| | - Hiroshi Handa
- Department of Nanoparticle Translational Research, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo 160-8402, Japan
| | - Makoto Suematsu
- Department of Biochemistry, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| |
Collapse
|
25
|
Sild M, Ruthazer ES, Booij L. Major depressive disorder and anxiety disorders from the glial perspective: Etiological mechanisms, intervention and monitoring. Neurosci Biobehav Rev 2017; 83:474-488. [DOI: 10.1016/j.neubiorev.2017.09.014] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 08/08/2017] [Accepted: 09/11/2017] [Indexed: 12/12/2022]
|
26
|
Decreased Motor Neuron Support by SMA Astrocytes due to Diminished MCP1 Secretion. J Neurosci 2017; 37:5309-5318. [PMID: 28450545 DOI: 10.1523/jneurosci.3472-16.2017] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Revised: 04/07/2017] [Accepted: 04/19/2017] [Indexed: 12/22/2022] Open
Abstract
Spinal muscular atrophy (SMA) is an autosomal-recessive disorder characterized by severe, often fatal muscle weakness due to loss of motor neurons. SMA patients have deletions and other mutations of the survival of motor neuron 1 (SMN1) gene, resulting in decreased SMN protein. Astrocytes are the primary support cells of the CNS and are responsible for glutamate clearance, metabolic support, response to injury, and regulation of signal transmission. Astrocytes have been implicated in SMA as in in other neurodegenerative disorders. Astrocyte-specific rescue of SMN protein levels has been shown to mitigate disease manifestations in mice. However, the mechanism by which SMN deficiency in astrocytes may contribute to SMA is unclear and what aspect of astrocyte activity is lacking is unknown. Therefore, it is worthwhile to identify defects in SMN-deficient astrocytes that compromise normal function. We show here that SMA astrocyte cultures derived from mouse spinal cord of both sexes are deficient in supporting both WT and SMN-deficient motor neurons derived from male, female, and mixed-sex sources and that this deficiency may be mitigated with secreted factors. In particular, SMN-deficient astrocytes have decreased levels of monocyte chemoactive protein 1 (MCP1) secretion compared with controls and MCP1 restoration stimulates outgrowth of neurites from cultured motor neurons. Correction of MCP1 deficiency may thus be a new therapeutic approach to SMA.SIGNIFICANCE STATEMENT Spinal muscular atrophy (SMA) is caused by the loss of motor neurons, but astrocyte dysfunction also contributes to the disease in mouse models. Monocyte chemoactive protein 1 (MCP1) has been shown to be neuroprotective and is released by astrocytes. Here, we report that MCP1 levels are decreased in SMA mice and that replacement of deficient MCP1 increases differentiation and neurite length of WT and SMN-deficient motor-neuron-like cells in cell culture. This study reveals a novel aspect of astrocyte dysfunction in SMA and indicates a possible approach for improving motor neuron growth and survival in this disease.
Collapse
|
27
|
Cahill MA, Jazayeri JA, Catalano SM, Toyokuni S, Kovacevic Z, Richardson DR. The emerging role of progesterone receptor membrane component 1 (PGRMC1) in cancer biology. Biochim Biophys Acta Rev Cancer 2016; 1866:339-349. [PMID: 27452206 DOI: 10.1016/j.bbcan.2016.07.004] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2016] [Revised: 07/17/2016] [Accepted: 07/19/2016] [Indexed: 01/09/2023]
Abstract
Progesterone receptor membrane component 1 (PGRMC1) is a multi-functional protein with a heme-binding moiety related to that of cytochrome b5, which is a putative progesterone receptor. The recently solved PGRMC1 structure revealed that heme-binding involves coordination by a tyrosinate ion at Y113, and induces dimerization which is stabilized by hydrophobic stacking of heme on adjacent monomers. Dimerization is required for association with cytochrome P450 (cyP450) enzymes, which mediates chemoresistance to doxorubicin and may be responsible for PGRMC1's anti-apoptotic activity. Here we review the multiple attested involvement of PGRMC1 in diverse functions, including regulation of cytochrome P450, steroidogenesis, vesicle trafficking, progesterone signaling and mitotic spindle and cell cycle regulation. Its wide range of biological functions is attested to particularly by its emerging association with cancer and progesterone-responsive female reproductive tissues. PGRMC1 exhibits all the hallmarks of a higher order nexus signal integration hub protein. It appears capable of acting as a detector that integrates information from kinase/phosphatase pathways with heme and CO levels and probably redox status.
Collapse
Affiliation(s)
- Michael A Cahill
- School of Biomedical Sciences, Charles Sturt University, Wagga Wagga, NSW 2678, Australia.
| | - Jalal A Jazayeri
- School of Biomedical Sciences, Charles Sturt University, Wagga Wagga, NSW 2678, Australia
| | - Susan M Catalano
- Cognition Therapeutics Inc., Pittsburgh, PA 15203, United States
| | - Shinya Toyokuni
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Japan
| | - Zaklina Kovacevic
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, New South Wales 2006, Australia
| | - Des R Richardson
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, New South Wales 2006, Australia.
| |
Collapse
|