1
|
Augusto-Oliveira M, Tremblay MÈ, Verkhratsky A. Receptors on Microglia. ADVANCES IN NEUROBIOLOGY 2024; 37:83-121. [PMID: 39207688 DOI: 10.1007/978-3-031-55529-9_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Microglial cells are the most receptive cells in the central nervous system (CNS), expressing several classes of receptors reflecting their immune heritage and newly acquired neural specialisation. Microglia possess, depending on the particular context, receptors to neurotransmitters and neuromodulators as well as immunocompetent receptors. This rich complement allows microglial cells to monitor the functional status of the nervous system, contribute actively to the regulation of neural activity and plasticity and homeostasis, and guard against pathogens as well as other challenges to the CNS's integrity and function.
Collapse
Affiliation(s)
- Marcus Augusto-Oliveira
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, Brazil
- Programa de Pós-Graduação em Farmacologia e Bioquímica, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, Brazil
| | - Marie-Ève Tremblay
- Division of Medical Sciences, Medical Sciences Building, University of Victoria, Victoria, BC, Canada
- Axe neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec City, QC, Canada
- Neurology and Neurosurgery Department, McGill University, Montreal, QC, Canada
- Department of Molecular Medicine, Université Laval, Pavillon Ferdinand-Vandry, Québec City, QC, Canada
- Department of Biochemistry and Molecular Biology, The University of British Columbia, Life Sciences Center, Vancouver, BC, Canada
| | - Alexei Verkhratsky
- Faculty of Life Sciences, The University of Manchester, Manchester, UK.
- Department of Neurosciences, University of the Basque Country, Leioa, Spain.
- IKERBASQUE, Basque Foundation for Science, Bilbao, Spain.
| |
Collapse
|
2
|
Ganesh T. Targeting EP2 Receptor for Drug Discovery: Strengths, Weaknesses, Opportunities, and Threats (SWOT) Analysis. J Med Chem 2023; 66:9313-9324. [PMID: 37458373 PMCID: PMC10388357 DOI: 10.1021/acs.jmedchem.3c00655] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Indexed: 07/28/2023]
Abstract
Cyclooxygenase-1 and -2 (COX1 and COX2) derived endogenous ligand prostaglandin-E2 (PGE2) triggers several physiological and pathological conditions. It mediates signaling through four G-protein coupled receptors, EP1, EP2, EP3, and EP4. Among these, EP2 is expressed throughout the body including the brain and uterus. The functional role of EP2 has been extensively studied using EP2 gene knockout mice, cellular models, and selective small molecule agonists and antagonists for this receptor. The efficacy data from in vitro and in vivo animal models indicate that EP2 receptor is a major proinflammatory mediator with deleterious functions in a variety of diseases suggesting a path forward for EP2 inhibitors as the next generation of selective anti-inflammatory and antiproliferative agents. Interestingly in certain diseases, EP2 action is beneficial; therefore, EP2 agonists seem to be clinically useful. Here, we highlight the strengths, weaknesses, opportunities, and potential threats (SWOT analysis) for targeting EP2 receptor for therapeutic development for a variety of unmet clinical needs.
Collapse
Affiliation(s)
- Thota Ganesh
- Department of Pharmacology and Chemical
Biology, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| |
Collapse
|
3
|
Pang QQ, Lee S, Cho EJ, Kim JH. Protective Effects of Cirsium japonicum var. maackii Flower on Amyloid Beta 25-35-Treated C6 Glial Cells. Life (Basel) 2023; 13:1453. [PMID: 37511827 PMCID: PMC10381248 DOI: 10.3390/life13071453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 06/09/2023] [Accepted: 06/23/2023] [Indexed: 07/30/2023] Open
Abstract
Amyloid beta (Aβ) is a neurotoxic peptide and a key factor causing Alzheimer's disease. Cirsium japonicum var. maackii (CJM) has neuroprotective effects, but the protective effects of the flower from CJM (FCJM) on the neural system remain unclear. This study aimed to identify the fraction of FCJM with the highest neuroprotective potential and investigate its protective mechanisms against Aβ25-35-induced inflammation in C6 glial cells. The cell viability and generation of reactive oxygen species (ROS) were measured to investigate the positive effect of FCJM on oxidative stress. Treatment with the FCJM extract or fractions increased the cell viability to 60-70% compared with 52% in the Aβ25-35-treated control group and decreased ROS production to 84% compared with 100% in the control group. The ethyl acetate fraction of FCJM (EFCJM) was the most effective among all the extracts and fractions. We analyzed the protective mechanisms of EFCJM on Aβ25-35-induced inflammation in C6 glial cells using Western blot. EFCJM downregulated amyloidogenic pathway-related proteins, such as Aβ precursor protein, β-secretase, presenilin 1, and presenilin 2. Moreover, EFCJM attenuated the Bax/Bcl-2 ratio, an index of apoptosis, and upregulated the oxidative stress-related protein, heme oxygenase-1. Therefore, this study demonstrated that FCJM improves cell viability and inhibits ROS in Aβ25-35-treated C6 glial cells. Furthermore, EFCJM exhibits neuroprotective effects in Aβ25-35-induced inflammation in C6 glial cells by modulating oxidative stress and amyloidogenic and apoptosis signaling pathways. FCJM, especially EFCJM, can be a promising agent for neurodegenerative disease prevention.
Collapse
Affiliation(s)
- Qi Qi Pang
- Department of Food Science and Nutrition, Kimchi Research Institute, Pusan National University, Busan 46241, Republic of Korea
| | - Sanghyun Lee
- Department of Plant Science and Technology, Chung-Ang University, Anseong 17546, Republic of Korea
- Natural Product Institute of Science and Technology, Anseong 17546, Republic of Korea
| | - Eun Ju Cho
- Department of Food Science and Nutrition, Kimchi Research Institute, Pusan National University, Busan 46241, Republic of Korea
| | - Ji-Hyun Kim
- Department of Food Science and Nutrition, Kimchi Research Institute, Pusan National University, Busan 46241, Republic of Korea
| |
Collapse
|
4
|
Monzón-Sandoval J, Burlacu E, Agarwal D, Handel AE, Wei L, Davis J, Cowley SA, Cader MZ, Webber C. Lipopolysaccharide distinctively alters human microglia transcriptomes to resemble microglia from Alzheimer's disease mouse models. Dis Model Mech 2022; 15:277958. [PMID: 36254682 PMCID: PMC9612871 DOI: 10.1242/dmm.049349] [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: 10/19/2021] [Accepted: 08/30/2022] [Indexed: 01/15/2023] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia, and risk-influencing genetics implicates microglia and neuroimmunity in the pathogenesis of AD. Induced pluripotent stem cell (iPSC)-derived microglia (iPSC-microglia) are increasingly used as a model of AD, but the relevance of historical immune stimuli to model AD is unclear. We performed a detailed cross-comparison over time on the effects of combinatory stimulation of iPSC-microglia, and in particular their relevance to AD. We used single-cell RNA sequencing to measure the transcriptional response of iPSC-microglia after 24 h and 48 h of stimulation with prostaglandin E2 (PGE2) or lipopolysaccharide (LPS)+interferon gamma (IFN-γ), either alone or in combination with ATPγS. We observed a shared core transcriptional response of iPSC-microglia to ATPγS and to LPS+IFN-γ, suggestive of a convergent mechanism of action. Across all conditions, we observed a significant overlap, although directional inconsistency to genes that change their expression levels in human microglia from AD patients. Using a data-led approach, we identify a common axis of transcriptomic change across AD genetic mouse models of microglia and show that only LPS provokes a transcriptional response along this axis in mouse microglia and LPS+IFN-γ in human iPSC-microglia. This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
| | - Elena Burlacu
- John Radcliffe Hospital, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DS, UK
| | - Devika Agarwal
- Nuffield Department of Medicine Research Building, Alzheimer's Research UK Oxford Drug Discovery Institute, University of Oxford, Oxford OX3 7FZ, UK.,Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Adam E Handel
- John Radcliffe Hospital, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DS, UK
| | - Liting Wei
- James Martin Stem Cell Facility, Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK
| | - John Davis
- Nuffield Department of Medicine Research Building, Alzheimer's Research UK Oxford Drug Discovery Institute, University of Oxford, Oxford OX3 7FZ, UK
| | - Sally A Cowley
- Translational Molecular Neuroscience Group, New Biochemistry Building, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX1 3QU, UK
| | - M Zameel Cader
- John Radcliffe Hospital, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DS, UK.,James Martin Stem Cell Facility, Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK
| | - Caleb Webber
- UK Dementia Research Institute, Cardiff University, Cardiff CF24 4HQ, UK
| |
Collapse
|
5
|
Rawat V, Banik A, Amaradhi R, Rojas A, Taval S, Nagy T, Dingledine R, Ganesh T. Pharmacological antagonism of EP2 receptor does not modify basal cardiovascular and respiratory function, blood cell counts, and bone morphology in animal models. Biomed Pharmacother 2022; 147:112646. [PMID: 35091236 PMCID: PMC8854338 DOI: 10.1016/j.biopha.2022.112646] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 12/31/2021] [Accepted: 01/12/2022] [Indexed: 01/12/2023] Open
Abstract
The EP2 receptor has emerged as a therapeutic target with exacerbating role in disease pathology for a variety of peripheral and central nervous system disorders. We and others have recently demonstrated beneficial effects of EP2 antagonists in preclinical models of neuroinflammation and peripheral inflammation. However, it was earlier reported that mice with global EP2 knockout (KO) display adverse phenotypes on fertility and blood pressure. Other studies indicated that EP2 activation with an agonist has a beneficial effect of healing fractured bone in animal models. These results impeded the development of EP2 antagonists, and EP2 antagonism as therapeutic strategy. To determine whether treatment with EP2 antagonist mimics the adverse phenotypes of the EP2 global KO mouse, we tested two EP2 antagonists TG11-77. HCl and TG6-10-1 in mice and rats while they are on normal or high-salt diet, and by two different administration protocols (acute and chronic). There were no adverse effects of the antagonists on systolic and diastolic blood pressure, heart rate, respiratory function in mice and rats regardless of rodents being on a regular or high salt diet. Furthermore, chronic exposure to TG11-77. HCl produced no adverse effects on blood cell counts, bone-volume and bone-mineral density in mice. Our findings argue against adverse effects on cardiovascular and respiratory systems, blood counts and bone structure in healthy rodents from the use of small molecule reversible antagonists for EP2, in contrast to the genetic ablation model. This study paves the way for advancing therapeutic applications of EP2 antagonists against diseases involving EP2 dysfunction.
Collapse
Affiliation(s)
- Varun Rawat
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| | - Avijit Banik
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| | - Radhika Amaradhi
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| | - Asheebo Rojas
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| | | | - Tamas Nagy
- Department of Pathology, College of Veterinary Medicine, University of Georgia, Athens GA 30602
| | - Raymond Dingledine
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| | - Thota Ganesh
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA 30322, USA.
| |
Collapse
|
6
|
Huang YC, Hsu SM, Shie FS, Shiao YJ, Chao LJ, Chen HW, Yao HH, Chien MA, Lin CC, Tsay HJ. Reduced mitochondria membrane potential and lysosomal acidification are associated with decreased oligomeric Aβ degradation induced by hyperglycemia: A study of mixed glia cultures. PLoS One 2022; 17:e0260966. [PMID: 35073330 PMCID: PMC8786178 DOI: 10.1371/journal.pone.0260966] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 11/20/2021] [Indexed: 01/21/2023] Open
Abstract
Diabetes is a risk factor for Alzheimer’s disease (AD), a chronic neurodegenerative disease. We and others have shown prediabetes, including hyperglycemia and obesity induced by high fat and high sucrose diets, is associated with exacerbated amyloid beta (Aβ) accumulation and cognitive impairment in AD transgenic mice. However, whether hyperglycemia reduce glial clearance of oligomeric amyloid-β (oAβ), the most neurotoxic Aβ aggregate, remains unclear. Mixed glial cultures simulating the coexistence of astrocytes and microglia in the neural microenvironment were established to investigate glial clearance of oAβ under normoglycemia and chronic hyperglycemia. Ramified microglia and low IL-1β release were observed in mixed glia cultures. In contrast, amoeboid-like microglia and higher IL-1β release were observed in primary microglia cultures. APPswe/PS1dE9 transgenic mice are a commonly used AD mouse model. Microglia close to senile plaques in APPswe/PS1dE9 transgenic mice exposed to normoglycemia or chronic hyperglycemia exhibited an amoeboid-like morphology; other microglia were ramified. Therefore, mixed glia cultures reproduce the in vivo ramified microglial morphology. To investigate the impact of sustained high-glucose conditions on glial oAβ clearance, mixed glia were cultured in media containing 5.5 mM glucose (normal glucose, NG) or 25 mM glucose (high glucose, HG) for 16 days. Compared to NG, HG reduced the steady-state level of oAβ puncta internalized by microglia and astrocytes and decreased oAβ degradation kinetics. Furthermore, the lysosomal acidification and lysosomal hydrolysis activity of microglia and astrocytes were lower in HG with and without oAβ treatment than NG. Moreover, HG reduced mitochondrial membrane potential and ATP levels in mixed glia, which can lead to reduced lysosomal function. Overall, continuous high glucose reduces microglial and astrocytic ATP production and lysosome activity which may lead to decreased glial oAβ degradation. Our study reveals diabetes-induced hyperglycemia hinders glial oAβ clearance and contributes to oAβ accumulation in AD pathogenesis.
Collapse
Affiliation(s)
- Yung-Cheng Huang
- Department of Physical Medicine and Rehabilitation, Cheng-Hsin General Hospital, Taipei, Taiwan, Republic of China
- National Taipei University of Nursing and Health Sciences, Taipei City, Taiwan, R.O.C
| | - Shu-Meng Hsu
- Institute of Neuroscience, School of Life Science, National Yang Ming Chiao Tung University, Taipei, Taiwan, R.O.C
| | - Feng-Shiun Shie
- Center for Neuropsychiatric Research National Health Research Institutes, Zhunan Town, Miaoli County, Taiwan, R.O.C
| | - Young-Ji Shiao
- National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei, Taiwan
- Ph.D. Program in Clinical Drug Development of Chinese Herbal Medicine, Taipei Medical University, Taipei, Taiwan, R.O.C
- Institute of Biopharmaceutical Science, National Yang Ming Chiao Tung University, Taipei, Taiwan, R.O.C
| | - Li-Jung Chao
- Institute of Neuroscience, School of Life Science, National Yang Ming Chiao Tung University, Taipei, Taiwan, R.O.C
| | - Hui-Wen Chen
- Institute of Neuroscience, School of Life Science, National Yang Ming Chiao Tung University, Taipei, Taiwan, R.O.C
| | - Heng-Hsiang Yao
- Institute of Neuroscience, School of Life Science, National Yang Ming Chiao Tung University, Taipei, Taiwan, R.O.C
| | - Meng An Chien
- Institute of Neuroscience, School of Life Science, National Yang Ming Chiao Tung University, Taipei, Taiwan, R.O.C
| | - Chung-Chih Lin
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan, Republic of China
- Brain Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan, Republic of China
- Biophotonics Interdisciplinary Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan, Republic of China
- * E-mail: (CCL); (HJT)
| | - Huey-Jen Tsay
- Institute of Neuroscience, School of Life Science, National Yang Ming Chiao Tung University, Taipei, Taiwan, R.O.C
- Brain Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan, Republic of China
- * E-mail: (CCL); (HJT)
| |
Collapse
|
7
|
Banik A, Amaradhi R, Lee D, Sau M, Wang W, Dingledine R, Ganesh T. Prostaglandin EP2 receptor antagonist ameliorates neuroinflammation in a two-hit mouse model of Alzheimer's disease. J Neuroinflammation 2021; 18:273. [PMID: 34801055 PMCID: PMC8605573 DOI: 10.1186/s12974-021-02297-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 10/14/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Alzheimer's disease (AD) causes substantial medical and societal burden with no therapies ameliorating cognitive deficits. Centralized pathologies involving amyloids, neurofibrillary tangles, and neuroinflammatory pathways are being investigated to identify disease-modifying targets for AD. Cyclooxygenase-2 (COX-2) is one of the potential neuroinflammatory agents involved in AD progression. However, chronic use of COX-2 inhibitors in patients produced adverse cardiovascular effects. We asked whether inhibition of EP2 receptors, downstream of the COX-2 signaling pathway, can ameliorate neuroinflammation in AD brains in presence or absence of a secondary inflammatory stimuli. METHODS We treated 5xFAD mice and their non-transgenic (nTg) littermates in presence or absence of lipopolysaccharide (LPS) with an EP2 antagonist (TG11-77.HCl). In cohort 1, nTg (no-hit) or 5xFAD (single-hit-genetic) mice were treated with vehicle or TG11-77.HCl for 12 weeks. In cohort 2, nTg (single-hit-environmental) and 5xFAD mice (two-hit) were administered LPS (0.5 mg/kg/week) and treated with vehicle or TG11-77.HCl for 8 weeks. RESULTS Complete blood count analysis showed that LPS induced anemia of inflammation in both groups in cohort 2. There was no adverse effect of LPS or EP2 antagonist on body weight throughout the treatment. In the neocortex isolated from the two-hit cohort of females, but not males, the elevated mRNA levels of proinflammatory mediators (IL-1β, TNF, IL-6, CCL2, EP2), glial markers (IBA1, GFAP, CD11b, S110B), and glial proteins were significantly reduced by EP2 antagonist treatment. Intriguingly, the EP2 antagonist had no effect on either of the single-hit cohorts. There was a modest increase in amyloid-plaque deposition upon EP2 antagonist treatment in the two-hit female brains, but not in the single-hit genetic female cohort. CONCLUSION These results reveal a potential neuroinflammatory role for EP2 in the two-hit 5xFAD mouse model. A selective EP2 antagonist reduces inflammation only in female AD mice subjected to a second inflammatory insult.
Collapse
Affiliation(s)
- Avijit Banik
- Department of Pharmacology and Chemical Biology, School of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Radhika Amaradhi
- Department of Pharmacology and Chemical Biology, School of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Daniel Lee
- Department of Pharmacology and Chemical Biology, School of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Michael Sau
- Department of Pharmacology and Chemical Biology, School of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Wenyi Wang
- Department of Pharmacology and Chemical Biology, School of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Raymond Dingledine
- Department of Pharmacology and Chemical Biology, School of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Thota Ganesh
- Department of Pharmacology and Chemical Biology, School of Medicine, Emory University, Atlanta, GA, 30322, USA.
| |
Collapse
|
8
|
Iwata Y, Miyao M, Hirotsu A, Tatsumi K, Matsuyama T, Uetsuki N, Tanaka T. The inhibitory effects of Orengedokuto on inducible PGE2 production in BV-2 microglial cells. Heliyon 2021; 7:e07759. [PMID: 34458607 PMCID: PMC8377439 DOI: 10.1016/j.heliyon.2021.e07759] [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: 05/31/2021] [Revised: 07/07/2021] [Accepted: 08/09/2021] [Indexed: 12/02/2022] Open
Abstract
Background and aim Reactive microglia has been associated with neuroinflammation caused by the production of proinflammatory molecules such as cytokines, nitric oxide, and prostaglandins. The overexpression of these molecules may provoke neuronal damage that can cause neurodegenerative diseases. A traditional herbal medicine, Orengedokuto (OGT), has been widely used for treating inflammation-related diseases. However, how it influences neuroinflammation remains poorly understood. Experimental procedure This study investigated the effects of OGT on inflammatory molecule induction in BV-2 microglial cells using real-time RT-PCR and ELISA. An in vivo confirmation of these effects was then performed in mice. Results and conclusion OGT showed dose-dependent inhibition of prostaglandin E2 (PGE2) production in BV-2 cells stimulated with lipopolysaccharide (LPS). To elucidate the mechanism of PGE2 inhibition, we examined cyclooxygenases (COXs) and found that OGT did not suppress COX-1 expression or inhibit LPS-induced COX-2 upregulation at either the transcriptional or translational levels. In addition, OGT did not inhibit COX enzyme activities within the concentration that inhibited PGE2 production, suggesting that the effect of OGT is COX-independent. The inhibitory effects of OGT on PGE2 production in BV-2 cells were experimentally replicated in primary cultured astrocytes and mice brains. OGT can be useful in the treatment of neuroinflammatory diseases by modulating PGE2 expression.
Collapse
Affiliation(s)
- Yoshika Iwata
- Department of Anesthesia, Kyoto University Hospital, 54 Kawahara-Cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Mariko Miyao
- Department of Anesthesia, Kyoto University Hospital, 54 Kawahara-Cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Akiko Hirotsu
- Department of Anesthesia, Kyoto University Hospital, 54 Kawahara-Cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Kenichiro Tatsumi
- Department of Anesthesia, Kyoto University Hospital, 54 Kawahara-Cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Tomonori Matsuyama
- Department of Anesthesia, National Hospital Organization Kyoto Medical Center, 1-1 Mukaihata-cho, Fukakusa, Fushimi-ku, Kyoto, 612-0861, Japan
| | - Nobuo Uetsuki
- Department of Anesthesia, Kyoto University Hospital, 54 Kawahara-Cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Tomoharu Tanaka
- Department of Anesthesia, Kyoto University Hospital, 54 Kawahara-Cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| |
Collapse
|
9
|
David S, López-Vales R. Bioactive Lipid Mediators in the Initiation and Resolution of Inflammation after Spinal Cord Injury. Neuroscience 2021; 466:273-297. [PMID: 33951502 DOI: 10.1016/j.neuroscience.2021.04.026] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 04/22/2021] [Accepted: 04/23/2021] [Indexed: 12/12/2022]
Abstract
Neuroinflammation is a prominent feature of the response to CNS trauma. It is also an important hallmark of various neurodegenerative diseases in which inflammation contributes to the progression of pathology. Inflammation in the CNS can contribute to secondary damage and is therefore an excellent therapeutic target for a range of neurological conditions. Inflammation in the nervous system is complex and varies in its fine details in different conditions. It involves a wide variety of secreted factors such as chemokines and cytokines, cell adhesion molecules, and different cell types that include resident cell of the CNS, as well as immune cells recruited from the peripheral circulation. Added to this complexity is the fact that some aspects of inflammation are beneficial, while other aspects can induce secondary damage in the acute, subacute and chronic phases. Understanding these aspects of the inflammatory profile is essential for developing effective therapies. Bioactive lipids constitute a large group of molecules that modulate the initiation and the resolution of inflammation. Dysregulation of these bioactive lipid pathways can lead to excessive acute inflammation, and failure to resolve this by specialized pro-resolution lipid mediators can lead to the development of chronic inflammation. The focus of this review is to discuss the effects of bioactive lipids in spinal cord trauma and their potential for therapies.
Collapse
Affiliation(s)
- Samuel David
- Centre for Research in Neuroscience, BRaIN Program, The Research Institute of the McGill University Health Centre, 1650 Cedar Avenue, Montreal, Quebec H3G 1A4, Canada.
| | - Rubén López-Vales
- Departament de Biologia Cellular, Fisiologia i Inmunologia, Institut de Neurociències, Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Universitat Autònoma de Barcelona, 08193 Bellaterra, Catalonia, Spain
| |
Collapse
|
10
|
Gorica E, Calderone V. Arachidonic Acid Derivatives and Neuroinflammation. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2021; 21:118-129. [PMID: 33557740 DOI: 10.2174/1871527320666210208130412] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 07/29/2020] [Accepted: 09/29/2020] [Indexed: 11/22/2022]
Abstract
Neuroinflammation is characterized by dysregulated inflammatory responses localized within the brain and spinal cord. Neuroinflammation plays a pivotal role in the onset of several neurodegenerative disorders and is considered a typical feature of these disorders. Microglia perform primary immune surveillance and macrophage-like activities within the central nervous system. Activated microglia are predominant players in the central nervous system response to damage related to stroke, trauma, and infection. Moreover, microglial activation per se leads to a proinflammatory response and oxidative stress. During the release of cytokines and chemokines, cyclooxygenases and phospholipase A2 are stimulated. Elevated levels of these compounds play a significant role in immune cell recruitment into the brain. Cyclic phospholipase A2 plays a fundamental role in the production of prostaglandins by releasing arachidonic acid. In turn, arachidonic acid is biotransformed through different routes into several mediators that are endowed with pivotal roles in the regulation of inflammatory processes. Some experimental models of neuroinflammation exhibit an increase in cyclic phospholipase A2, leukotrienes, and prostaglandins such as prostaglandin E2, prostaglandin D2, or prostacyclin. However, findings on the role of the prostacyclin receptors have revealed that their signalling suppresses Th2-mediated inflammatory responses. In addition, other in vitro evidence suggests that prostaglandin E2 may inhibit the production of some inflammatory cytokines, attenuating inflammatory events such as mast cell degranulation or inflammatory leukotriene production. Based on these conflicting experimental data, the role of arachidonic acid derivatives in neuroinflammation remains a challenging issue.
Collapse
Affiliation(s)
- Era Gorica
- Department of Pharmacy, University of Pisa, Pisa. Italy
| | | |
Collapse
|
11
|
Montecillo-Aguado M, Tirado-Rodriguez B, Tong Z, Vega OM, Morales-Martínez M, Abkenari S, Pedraza-Chaverri J, Huerta-Yepez S. Importance of the Role of ω-3 and ω-6 Polyunsaturated Fatty Acids in the Progression of Brain Cancer. Brain Sci 2020; 10:E381. [PMID: 32560280 PMCID: PMC7349634 DOI: 10.3390/brainsci10060381] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 05/28/2020] [Accepted: 06/09/2020] [Indexed: 12/21/2022] Open
Abstract
Brain cancer is one of the most malignant types of cancer in both children and adults. Brain cancer patients tend to have a poor prognosis and a high rate of mortality. Additionally, 20-40% of all other types of cancer can develop brain metastasis. Numerous pieces of evidence suggest that omega-3-polyunsaturated fatty acids (ω-PUFAs) could potentially be used in the prevention and therapy of several types of cancer. PUFAs and oxylipins are fundamental in preserving physiological events in the nervous system; it is, therefore, necessary to maintain a certain ratio of ω-3 to ω-6 for normal nervous system function. Alterations in PUFAs signaling are involved in the development of various pathologies of the nervous system, including cancer. It is well established that an omega-6-polyunsaturated fatty acid (ω-6 PUFA)-rich diet has a pro-tumoral effect, whereas the consumption of an ω-3 rich diet has an anti-tumoral effect. This review aims to offer a better understanding of brain cancer and PUFAs and to discuss the role and impact of PUFAs on the development of different types of brain cancer. Considering the difficulty of antitumor drugs in crossing the blood-brain barrier, the therapeutic role of ω-3/ω-6 PUFAs against brain cancer would be a good alternative to consider. We highlight our current understanding of the role of PUFAs and its metabolites (oxylipins) in different brain tumors, proliferation, apoptosis, invasion, angiogenesis, and immunosuppression by focusing on recent research in vitro and in vivo.
Collapse
Affiliation(s)
- Mayra Montecillo-Aguado
- Programa de Doctorado en Ciencias Biomédicas, Facultad de Medicina, Universidad Nacional Autónoma de Mexico (UNAM), Mexico City 04510, Mexico;
- Hospital Infantil de Mexico, Federico Gomez, Unidad de Investigacion en Enfermedades Oncologicas, Mexico City 06720, Mexico; (B.T.-R.); (M.M.-M.)
| | - Belen Tirado-Rodriguez
- Hospital Infantil de Mexico, Federico Gomez, Unidad de Investigacion en Enfermedades Oncologicas, Mexico City 06720, Mexico; (B.T.-R.); (M.M.-M.)
| | - Zhen Tong
- Molecular Toxicology Interdepartmental Program and Environmental Health Sciences, University of California, Los Angeles, CA 90095, USA;
- Department of Pathology & Laboratory Medicine, University of California, Los Angeles, CA 90095, USA; (O.M.V.); (S.A.)
| | - Owen M. Vega
- Department of Pathology & Laboratory Medicine, University of California, Los Angeles, CA 90095, USA; (O.M.V.); (S.A.)
| | - Mario Morales-Martínez
- Hospital Infantil de Mexico, Federico Gomez, Unidad de Investigacion en Enfermedades Oncologicas, Mexico City 06720, Mexico; (B.T.-R.); (M.M.-M.)
| | - Shaheen Abkenari
- Department of Pathology & Laboratory Medicine, University of California, Los Angeles, CA 90095, USA; (O.M.V.); (S.A.)
| | - José Pedraza-Chaverri
- Departamento de Biología, Facultad de Química, Universidad Nacional Autonoma de Mexico (UNAM), Mexico City 04510, Mexico;
| | - Sara Huerta-Yepez
- Hospital Infantil de Mexico, Federico Gomez, Unidad de Investigacion en Enfermedades Oncologicas, Mexico City 06720, Mexico; (B.T.-R.); (M.M.-M.)
- Department of Pathology & Laboratory Medicine, University of California, Los Angeles, CA 90095, USA; (O.M.V.); (S.A.)
| |
Collapse
|
12
|
Amaradhi R, Banik A, Mohammed S, Patro V, Rojas A, Wang W, Motati DR, Dingledine R, Ganesh T. Potent, Selective, Water Soluble, Brain-Permeable EP2 Receptor Antagonist for Use in Central Nervous System Disease Models. J Med Chem 2020; 63:1032-1050. [PMID: 31904232 PMCID: PMC7394479 DOI: 10.1021/acs.jmedchem.9b01218] [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] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Activation of prostanoid EP2 receptor exacerbates neuroinflammatory and neurodegenerative pathology in central nervous system diseases such as epilepsy, Alzheimer's disease, and cerebral aneurysms. A selective and brain-permeable EP2 antagonist will be useful to attenuate the inflammatory consequences of EP2 activation and to reduce the severity of these chronic diseases. We recently developed a brain-permeable EP2 antagonist 1 (TG6-10-1), which displayed anti-inflammatory and neuroprotective actions in rodent models of status epilepticus. However, this compound exhibited moderate selectivity to EP2, a short plasma half-life in rodents (1.7 h) and low aqueous solubility (27 μM), limiting its use in animal models of chronic disease. With lead-optimization studies, we have developed several novel EP2 antagonists with improved water solubility, brain penetration, high EP2 potency, and selectivity. These novel inhibitors suppress inflammatory gene expression induced by EP2 receptor activation in a microglial cell line, reinforcing the use of EP2 antagonists as anti-inflammatory agents.
Collapse
Affiliation(s)
- Radhika Amaradhi
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, 1510 Clifton Rd; Atlanta, GA, 30322, United States of America
| | - Avijit Banik
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, 1510 Clifton Rd; Atlanta, GA, 30322, United States of America
| | - Shabber Mohammed
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, 1510 Clifton Rd; Atlanta, GA, 30322, United States of America
| | - Vidyavathi Patro
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, 1510 Clifton Rd; Atlanta, GA, 30322, United States of America
| | - Asheebo Rojas
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, 1510 Clifton Rd; Atlanta, GA, 30322, United States of America
| | - Wenyi Wang
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, 1510 Clifton Rd; Atlanta, GA, 30322, United States of America
| | - Damoder Reddy Motati
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, 1510 Clifton Rd; Atlanta, GA, 30322, United States of America
| | - Ray Dingledine
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, 1510 Clifton Rd; Atlanta, GA, 30322, United States of America
| | - Thota Ganesh
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, 1510 Clifton Rd; Atlanta, GA, 30322, United States of America
| |
Collapse
|
13
|
Kays JS, Yamamoto BK. Evaluation of Microglia/Macrophage Cells from Rat Striatum and Prefrontal Cortex Reveals Differential Expression of Inflammatory-Related mRNA after Methamphetamine. Brain Sci 2019; 9:brainsci9120340. [PMID: 31775383 PMCID: PMC6955783 DOI: 10.3390/brainsci9120340] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 11/19/2019] [Accepted: 11/20/2019] [Indexed: 12/19/2022] Open
Abstract
RNA sequencing (RNAseq) can be a powerful tool in the identification of transcriptional changes after drug treatment. RNAseq was utilized to determine expression changes in Fluorescence-activated cell sorted (FACS) CD11b/c+ cells from the striatum (STR) and prefrontal cortex (PFC) of male Sprague-Dawley rats after a methamphetamine (METH) binge dosing regimen. Resident microglia and infiltrating macrophages were collected 2 h or 3 days after drug administration. Gene expression changes indicated there was an increase toward an overall pro-inflammatory state, or M1 polarization, along with what appears to be a subset of cells that differentiated toward the anti-inflammatory M2 polarization. In general, there were significantly more mRNA expression changes in the STR than the PFC and more at 2 h post-binge METH than at 3 days post-binge METH. Additionally, Ingenuity® Pathway Analysis along with details of RNA expression changes revealed cyclo-oxygenase 2 (COX2)-driven prostaglandin (PG) E2 synthesis, glutamine uptake, and the Nuclear factor erythroid2-related factor 2 (NRF2) canonical pathway in microglia were associated with the binge administration regimen of METH.
Collapse
|
14
|
Rojas A, Banik A, Chen D, Flood K, Ganesh T, Dingledine R. Novel Microglia Cell Line Expressing the Human EP2 Receptor. ACS Chem Neurosci 2019; 10:4280-4292. [PMID: 31469538 DOI: 10.1021/acschemneuro.9b00311] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Recently, EP2 signaling pathways were shown to regulate the classical activation and death of microglia in rat primary microglial culture. The study of microglial cells has been challenging because they are time-consuming to isolate in culture, they are demanding in their growth requirements, and they have a limited lifespan. To circumvent these difficulties, we created a murine BV2 microglial cell line stably expressing human EP2 receptors (BV2-hEP2) and further explored EP2 modulation of microglial functions. The BV2-hEP2 cells displayed cAMP elevation when exposed to the selective EP2 receptor agonists (ONO-AE1-259-1 and CP544326), and this response was competitively inhibited by TG4-155, a selective EP2 antagonist (Schild KB = 2.6 nM). By contrast, untransfected BV2 cells were unresponsive to selective EP2 agonists. Similar to the case of rat primary microglia, BV2-hEP2 microglia treated with lipopolysaccharide (LPS) (100 ng/mL) displayed rapid and robust induction of the inflammatory mediators COX-2, IL-1β, TNFα, and IL-6. EP2 activation depressed TNFα induction but exacerbated that of the other inflammatory mediators. Like primary microglia, classically activated BV2 microglia phagocytose fluorescent-labeled latex microspheres. The presence of EP2, but not its activation by agonists, in BV2-hEP2 microglia reduced phagocytosis and proliferation by 65% and 32%, respectively, compared to BV2 microglia. Thus, BV2-hEP2 is the first microglial cell line that retains the EP2 modulation of immune regulation and phagocytic ability of native microglia. Suppression of phagocytosis by the EP2 protein appears unrelated to classical EP2 signaling pathways, which has implications for therapeutic development of EP2 antagonists.
Collapse
Affiliation(s)
- Asheebo Rojas
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| | - Avijit Banik
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| | - Di Chen
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| | - Kevin Flood
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| | - Thota Ganesh
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| | - Raymond Dingledine
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| |
Collapse
|
15
|
Golia MT, Poggini S, Alboni S, Garofalo S, Ciano Albanese N, Viglione A, Ajmone-Cat MA, St-Pierre A, Brunello N, Limatola C, Branchi I, Maggi L. Interplay between inflammation and neural plasticity: Both immune activation and suppression impair LTP and BDNF expression. Brain Behav Immun 2019; 81:484-494. [PMID: 31279682 DOI: 10.1016/j.bbi.2019.07.003] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Revised: 06/29/2019] [Accepted: 07/03/2019] [Indexed: 01/11/2023] Open
Abstract
An increasing number of studies show that both inflammation and neural plasticity act as key players in the vulnerability and recovery from psychiatric disorders and neurodegenerative diseases. However, the interplay between these two players has been limitedly explored. In fact, while a few studies reported an immune activation, others conveyed an immune suppression, associated with an impairment in neural plasticity. Therefore, we hypothesized that deviations in inflammatory levels in both directions may impair neural plasticity. We tested this hypothesis experimentally, by acute treatment of C57BL/6 adult male mice with different doses of two inflammatory modulators: lipopolysaccharide (LPS), an endotoxin, and ibuprofen (IBU), a nonselective cyclooxygenase inhibitor, which are respectively a pro- and an anti-inflammatory agent. The results showed that LPS and IBU have different effects on behavior and inflammatory response. LPS treatment induced a reduction of body temperature, a decrease of body weight and a reduced food and liquid intake. In addition, it led to increased levels of inflammatory markers expression, both in the total hippocampus and in isolated microglia cells, including Interleukin (IL)-1β, and enhanced the concentration of prostaglandin E2 (PGE2). On the other hand, IBU increased the level of anti-inflammatory markers, decreased tryptophan 2,3-dioxygenase (TDO2), the first step in the kynurenine pathway known to be activated during inflammatory conditions, and PGE2 levels. Though LPS and IBU administration differently affected mediators related with pro- or anti-inflammatory responses, they produced overlapping effects on neural plasticity. Indeed, higher doses of both LPS and IBU induced a statistically significant decrease in the amplitude of long-term potentiation (LTP), in Brain-Derived Neurotrophic Factor (BDNF) expression levels and in the phosphorylation of the AMPA (α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid) receptor subunit GluR1, compared to the control group. Such effect appears to be dose-dependent since only the higher, but not the lower, dose of both compounds led to a plasticity impairment. Overall, the present findings indicate that acute treatment with pro- and anti-inflammatory agents impair neural plasticity in a dose dependent manner.
Collapse
Affiliation(s)
- Maria Teresa Golia
- Department of Physiology and Pharmacology, Laboratory Affiliated to Istituto Pasteur-Italy, Sapienza University of Rome, Italy
| | - Silvia Poggini
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Rome, Italy
| | - Silvia Alboni
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Stefano Garofalo
- Department of Physiology and Pharmacology, Laboratory Affiliated to Istituto Pasteur-Italy, Sapienza University of Rome, Italy
| | - Naomi Ciano Albanese
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Rome, Italy
| | - Aurelia Viglione
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Rome, Italy; PhD Program in Neuroscience, Scuola Superiore di Pisa, Pisa, Italy
| | | | - Abygaël St-Pierre
- Axe Neurosciences, Centre de recherche du CHU de Québec, Québec, Canada
| | - Nicoletta Brunello
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Cristina Limatola
- Department of Physiology and Pharmacology, Laboratory Affiliated to Istituto Pasteur-Italy, Sapienza University of Rome, Italy; IRCCS Neuromed, Pozzilli, Isernia, Italy
| | - Igor Branchi
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Rome, Italy
| | - Laura Maggi
- Department of Physiology and Pharmacology, Laboratory Affiliated to Istituto Pasteur-Italy, Sapienza University of Rome, Italy.
| |
Collapse
|
16
|
Biringer RG. The Role of Eicosanoids in Alzheimer's Disease. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2019; 16:ijerph16142560. [PMID: 31323750 PMCID: PMC6678666 DOI: 10.3390/ijerph16142560] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 07/09/2019] [Accepted: 07/13/2019] [Indexed: 12/21/2022]
Abstract
Alzheimer's disease (AD) is one of the most common neurodegenerative disorders known. Estimates from the Alzheimer's Association suggest that there are currently 5.8 million Americans living with the disease and that this will rise to 14 million by 2050. Research over the decades has revealed that AD pathology is complex and involves a number of cellular processes. In addition to the well-studied amyloid-β and tau pathology, oxidative damage to lipids and inflammation are also intimately involved. One aspect all these processes share is eicosanoid signaling. Eicosanoids are derived from polyunsaturated fatty acids by enzymatic or non-enzymatic means and serve as short-lived autocrine or paracrine agents. Some of these eicosanoids serve to exacerbate AD pathology while others serve to remediate AD pathology. A thorough understanding of eicosanoid signaling is paramount for understanding the underlying mechanisms and developing potential treatments for AD. In this review, eicosanoid metabolism is examined in terms of in vivo production, sites of production, receptor signaling, non-AD biological functions, and known participation in AD pathology.
Collapse
Affiliation(s)
- Roger G Biringer
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, 5000 Lakewood Ranch Blvd., Bradenton, FL 34211, USA.
| |
Collapse
|
17
|
Hsiao HT, Lee YC, Liu YC, Kuo PC, Wu SN. Differential suppression of delayed-rectifier and inwardly rectifier K + currents by a group of ent-kaurane-type diterpenoids from Croton tonkinensis, in microglial cells. Eur J Pharmacol 2019; 856:172414. [PMID: 31129155 DOI: 10.1016/j.ejphar.2019.172414] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 05/14/2019] [Accepted: 05/22/2019] [Indexed: 12/11/2022]
Abstract
Croton is an extensive flowering plant genus in the spurge family, Euphorbiaceae. Three croton compounds with the common ent-kaurane skeleton were purified from Croton tonkinensis. By using patch-clamp recording technique, we thoroughly examined the effect of a group of croton compounds, croton-01 (ent-18-acetoxy-7α-hydroxykaur-16-en-15-one), croton-02 (ent-7α,14β-dihydroxykaur-16-en-15-one), and croton-03 (ent-1β-acetoxy-7α,14β-dihydroxykaur-16-en-15-one), on the membrane current in SM826 and BV2 microglial cells. Although neither voltage-gated Na+ nor Ca2+ currents were present in these cells, both delayed-rectifier K+ outward (IK(DR)) and inwardly rectifying K+ currents (IK(IR)) were readily detected. Croton-03 differentially caused inhibition of IK(DR) or IK(IR) in a concentration-dependent manner. According to a minimal scheme, the shortening of the time constant in either the IK(DR)-related block or IK(IR) caused by different concentrations of croton-03 was quantitatively estimated with a dissociation constant of 6.45 and 29.5 μM, respectively. In SM826 cells differentiated with β-amyloid, inhibitory action on these K+ currents remained unaltered. In ultraviolet C-irradiated cells, the magnitude of IK(IR) was still decreased by addition of croton-03. Therefore, our study suggests that these ent-kaurane diterpenoids ought to somehow act on the cellular mechanisms by which they influence the functional activities of microglial cells.
Collapse
Affiliation(s)
- Hung-Tsung Hsiao
- Department of Anesthesiology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan City, Taiwan
| | - Yu-Chi Lee
- Department of Physiology, National Cheng Kung University Medical College, Tainan City, Taiwan
| | - Yen-Chin Liu
- Department of Anesthesiology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan City, Taiwan
| | - Ping-Chung Kuo
- School of Pharmacy, National Cheng Kung University, Tainan City, Taiwan
| | - Sheng-Nan Wu
- Department of Physiology, National Cheng Kung University Medical College, Tainan City, Taiwan; Institute of Basic Medical Sciences, National Cheng Kung University Medical College, Tainan City, Taiwan.
| |
Collapse
|
18
|
Han J, Zhang M, Lin HY, Huang FY, Lin YY, Tan GH, Zheng ZY. Impaired Autophagic Degradation of Transforming Growth Factor-β-Induced Protein by Macrophages in Lattice Corneal Dystrophy. Invest Ophthalmol Vis Sci 2019; 60:978-989. [PMID: 30884524 DOI: 10.1167/iovs.18-25838] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose Lattice corneal dystrophy (LCD) is related to the denaturation of transforming growth factor-β-induced protein (TGFBIp). Autophagic degradation of the denatured proteins by macrophages is one pathway to remove the denatured proteins. Thus, we investigated the role of autophagy in the degradation of mutant (MU) TGFBIp in macrophages. Methods Corneas from participants were observed by slit-lamp photography and subjected to histopathologic and genetic analysis. Wild-type (WT) and MU TGFBIp were recombined and expressed. Macrophages from MU participants were isolated and cocultured with the recombinant TGFBIp. Colocalization of the two molecules was observed by immunofluorescent microscopy. Enzyme-linked immunosorbent assay, Western blotting, and flow cytometry were used to detect changes in molecule expression related to the phenotype and autophagy process. Results Fourteen members from a family of 25 were identified as LCD sufferers. Significant TGFBIp aggregates and macrophage infiltration were found only in the corneas of LCD sufferers. Marker accumulation of TGFBIp was found in macrophages exposed to MU TGFBIp even at 5 hours after MU TGFBIp was withdrawn. High expressions of CD68 and CD36 were found in macrophages exposed to WT TGFBIp, but not to MU TGFBIp. Impaired autophagic flux due to defective autophagosome fusion to lysosomes was found in macrophages exposed to MU TGFBIp. Blockage of the autophagic process suppressed the expression of CD68 and CD36 in macrophages exposed to WT TGFBIp to levels similar to those found in macrophages exposed to MU TGFBIp. Conclusions Our results suggested that reversion of the defective autophagic process in macrophages may be a therapeutic strategy for patients with LCD.
Collapse
Affiliation(s)
- Ji Han
- Hainan Provincial Key Laboratory of Tropical Medicine, Hainan Medical College, Haikou, China.,Department of Ophthalmology, The First Affiliated Hospital of Hainan Medical College, Haikou, China
| | - Ming Zhang
- Hainan Provincial Key Laboratory of Tropical Medicine, Hainan Medical College, Haikou, China
| | - Hai-Yan Lin
- Hainan Provincial Key Laboratory of Tropical Medicine, Hainan Medical College, Haikou, China
| | - Feng-Ying Huang
- Hainan Provincial Key Laboratory of Tropical Medicine, Hainan Medical College, Haikou, China
| | - Ying-Ying Lin
- Hainan Provincial Key Laboratory of Tropical Medicine, Hainan Medical College, Haikou, China
| | - Guang-Hong Tan
- Hainan Provincial Key Laboratory of Tropical Medicine, Hainan Medical College, Haikou, China
| | - Zhen-You Zheng
- Hainan Provincial Key Laboratory of Tropical Medicine, Hainan Medical College, Haikou, China.,Department of Ophthalmology, The First Affiliated Hospital of Hainan Medical College, Haikou, China
| |
Collapse
|
19
|
Zádori D, Veres G, Szalárdy L, Klivényi P, Vécsei L. Alzheimer's Disease: Recent Concepts on the Relation of Mitochondrial Disturbances, Excitotoxicity, Neuroinflammation, and Kynurenines. J Alzheimers Dis 2019; 62:523-547. [PMID: 29480191 DOI: 10.3233/jad-170929] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The pathomechanism of Alzheimer's disease (AD) certainly involves mitochondrial disturbances, glutamate excitotoxicity, and neuroinflammation. The three main aspects of mitochondrial dysfunction in AD, i.e., the defects in dynamics, altered bioenergetics, and the deficient transport, act synergistically. In addition, glutamatergic neurotransmission is affected in several ways. The balance between synaptic and extrasynaptic glutamatergic transmission is shifted toward the extrasynaptic site contributing to glutamate excitotoxicity, a phenomenon augmented by increased glutamate release and decreased glutamate uptake. Neuroinflammation in AD is predominantly linked to central players of the innate immune system, with central nervous system (CNS)-resident microglia, astroglia, and perivascular macrophages having been implicated at the cellular level. Several abnormalities have been described regarding the activation of certain steps of the kynurenine (KYN) pathway of tryptophan metabolism in AD. First of all, the activation of indolamine 2,3-dioxygenase, the first and rate-limiting step of the pathway, is well-demonstrated. 3-Hydroxy-L-KYN and its metabolite, 3-hydroxy-anthranilic acid have pro-oxidant, antioxidant, and potent immunomodulatory features, giving relevance to their alterations in AD. Another metabolite, quinolinic acid, has been demonstrated to be neurotoxic, promoting glutamate excitotoxicity, reactive oxygen species production, lipid peroxidation, and microglial neuroinflammation, and its abundant presence in AD pathologies has been demonstrated. Finally, the neuroprotective metabolite, kynurenic acid, has been associated with antagonistic effects at glutamate receptors, free radical scavenging, and immunomodulation, giving rise to potential therapeutic implications. This review presents the multiple connections of KYN pathway-related alterations to three main domains of AD pathomechanism, such as mitochondrial dysfunction, excitotoxicity, and neuroinflammation, implicating possible therapeutic options.
Collapse
Affiliation(s)
- Dénes Zádori
- Department of Neurology, Faculty of Medicine, Albert Szent-Györgyi Clinical Center, University of Szeged, Szeged, Hungary
| | - Gábor Veres
- Department of Neurology, Faculty of Medicine, Albert Szent-Györgyi Clinical Center, University of Szeged, Szeged, Hungary
| | - Levente Szalárdy
- Department of Neurology, Faculty of Medicine, Albert Szent-Györgyi Clinical Center, University of Szeged, Szeged, Hungary
| | - Péter Klivényi
- Department of Neurology, Faculty of Medicine, Albert Szent-Györgyi Clinical Center, University of Szeged, Szeged, Hungary
| | - László Vécsei
- Department of Neurology, Faculty of Medicine, Albert Szent-Györgyi Clinical Center, University of Szeged, Szeged, Hungary.,MTA-SZTE Neuroscience Research Group, Szeged, Hungary
| |
Collapse
|
20
|
Nazmi A, Field RH, Griffin EW, Haugh O, Hennessy E, Cox D, Reis R, Tortorelli L, Murray CL, Lopez-Rodriguez AB, Jin L, Lavelle EC, Dunne A, Cunningham C. Chronic neurodegeneration induces type I interferon synthesis via STING, shaping microglial phenotype and accelerating disease progression. Glia 2019; 67:1254-1276. [PMID: 30680794 PMCID: PMC6520218 DOI: 10.1002/glia.23592] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 12/21/2018] [Accepted: 12/28/2018] [Indexed: 12/13/2022]
Abstract
Type I interferons (IFN‐I) are the principal antiviral molecules of the innate immune system and can be made by most cell types, including central nervous system cells. IFN‐I has been implicated in neuroinflammation during neurodegeneration, but its mechanism of induction and its consequences remain unclear. In the current study, we assessed expression of IFN‐I in murine prion disease (ME7) and examined the contribution of the IFN‐I receptor IFNAR1 to disease progression. The data indicate a robust IFNβ response, specifically in microglia, with evidence of IFN‐dependent genes in both microglia and astrocytes. This IFN‐I response was absent in stimulator of interferon genes (STING−/−) mice. Microglia showed increased numbers and activated morphology independent of genotype, but transcriptional signatures indicated an IFNAR1‐dependent neuroinflammatory phenotype. Isolation of microglia and astrocytes demonstrated disease‐associated microglial induction of Tnfα, Tgfb1, and of phagolysosomal system transcripts including those for cathepsins, Cd68, C1qa, C3, and Trem2, which were diminished in IFNAR1 and STING deficient mice. Microglial increases in activated cathepsin D, and CD68 were significantly reduced in IFNAR1−/− mice, particularly in white matter, and increases in COX‐1 expression, and prostaglandin synthesis were significantly mitigated. Disease progressed more slowly in IFNAR1−/− mice, with diminished synaptic and neuronal loss and delayed onset of neurological signs and death but without effect on proteinase K‐resistant PrP levels. Therefore, STING‐dependent IFN‐I influences microglial phenotype and influences neurodegenerative progression despite occurring secondary to initial degenerative changes. These data expand our mechanistic understanding of IFN‐I induction and its impact on microglial function during chronic neurodegeneration.
Collapse
Affiliation(s)
- Arshed Nazmi
- School of Biochemistry and Immunology, Trinity College Institute of Neuroscience & Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Republic of Ireland
| | - Robert H Field
- School of Biochemistry and Immunology, Trinity College Institute of Neuroscience & Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Republic of Ireland
| | - Eadaoin W Griffin
- School of Biochemistry and Immunology, Trinity College Institute of Neuroscience & Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Republic of Ireland
| | - Orla Haugh
- School of Biochemistry and Immunology, Trinity College Institute of Neuroscience & Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Republic of Ireland
| | - Edel Hennessy
- School of Biochemistry and Immunology, Trinity College Institute of Neuroscience & Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Republic of Ireland
| | - Donal Cox
- School of Biochemistry and Immunology, Trinity College Institute of Neuroscience & Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Republic of Ireland
| | - Renata Reis
- School of Biochemistry and Immunology, Trinity College Institute of Neuroscience & Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Republic of Ireland
| | - Lucas Tortorelli
- School of Biochemistry and Immunology, Trinity College Institute of Neuroscience & Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Republic of Ireland
| | - Carol L Murray
- School of Biochemistry and Immunology, Trinity College Institute of Neuroscience & Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Republic of Ireland
| | - Ana Belen Lopez-Rodriguez
- School of Biochemistry and Immunology, Trinity College Institute of Neuroscience & Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Republic of Ireland
| | - Lei Jin
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, University of Florida, Gainesville, Florida
| | - Ed C Lavelle
- School of Biochemistry and Immunology, Trinity College Institute of Neuroscience & Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Republic of Ireland
| | - Aisling Dunne
- School of Biochemistry and Immunology, Trinity College Institute of Neuroscience & Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Republic of Ireland
| | - Colm Cunningham
- School of Biochemistry and Immunology, Trinity College Institute of Neuroscience & Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Republic of Ireland
| |
Collapse
|
21
|
Yao XC, Xue X, Zhang HT, Zhu MM, Yang XW, Wu CF, Yang JY. Pseudoginsenoside‐F11 alleviates oligomeric β‐amyloid‐induced endosome‐lysosome defects in microglia. Traffic 2018; 20:61-70. [DOI: 10.1111/tra.12620] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 10/26/2018] [Accepted: 10/26/2018] [Indexed: 12/13/2022]
Affiliation(s)
- Xue C. Yao
- Department of PharmacologyShenyang Pharmaceutical University Shenyang PR China
| | - Xue Xue
- State Key Laboratory of Medicinal Chemical Biology, College of PharmacyNankai University Tianjin PR China
| | - Hao T. Zhang
- Department of PharmacologyShenyang Pharmaceutical University Shenyang PR China
| | - Ming M. Zhu
- Department of PharmacologyShenyang Pharmaceutical University Shenyang PR China
| | - Xiao W. Yang
- Department of PharmacologyShenyang Pharmaceutical University Shenyang PR China
| | - Chun F. Wu
- Department of PharmacologyShenyang Pharmaceutical University Shenyang PR China
| | - Jing Y. Yang
- Department of PharmacologyShenyang Pharmaceutical University Shenyang PR China
| |
Collapse
|
22
|
Greenhalgh AD, Zarruk JG, Healy LM, Baskar Jesudasan SJ, Jhelum P, Salmon CK, Formanek A, Russo MV, Antel JP, McGavern DB, McColl BW, David S. Peripherally derived macrophages modulate microglial function to reduce inflammation after CNS injury. PLoS Biol 2018; 16:e2005264. [PMID: 30332405 PMCID: PMC6205650 DOI: 10.1371/journal.pbio.2005264] [Citation(s) in RCA: 148] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 10/29/2018] [Accepted: 10/01/2018] [Indexed: 12/19/2022] Open
Abstract
Infiltrating monocyte-derived macrophages (MDMs) and resident microglia dominate central nervous system (CNS) injury sites. Differential roles for these cell populations after injury are beginning to be uncovered. Here, we show evidence that MDMs and microglia directly communicate with one another and differentially modulate each other's functions. Importantly, microglia-mediated phagocytosis and inflammation are suppressed by infiltrating macrophages. In the context of spinal cord injury (SCI), preventing such communication increases microglial activation and worsens functional recovery. We suggest that macrophages entering the CNS provide a regulatory mechanism that controls acute and long-term microglia-mediated inflammation, which may drive damage in a variety of CNS conditions.
Collapse
Affiliation(s)
- Andrew D. Greenhalgh
- Centre for Research in Neuroscience, The Research Institute of the McGill University Health Center, Quebec, Canada
- Laboratory of Nutrition and Integrated Neurobiology, UMR INRA 1286, University of Bordeaux, Bordeaux, France
| | - Juan G. Zarruk
- Centre for Research in Neuroscience, The Research Institute of the McGill University Health Center, Quebec, Canada
| | - Luke M. Healy
- Neuroimmunology Unit, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Sam J. Baskar Jesudasan
- Centre for Research in Neuroscience, The Research Institute of the McGill University Health Center, Quebec, Canada
| | - Priya Jhelum
- Centre for Research in Neuroscience, The Research Institute of the McGill University Health Center, Quebec, Canada
| | - Christopher K. Salmon
- Centre for Research in Neuroscience, The Research Institute of the McGill University Health Center, Quebec, Canada
| | - Albert Formanek
- Centre for Research in Neuroscience, The Research Institute of the McGill University Health Center, Quebec, Canada
| | - Matthew V. Russo
- Viral Immunology and Intravital Imaging Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Jack P. Antel
- Neuroimmunology Unit, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Dorian B. McGavern
- Viral Immunology and Intravital Imaging Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Barry W. McColl
- UK Dementia Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Samuel David
- Centre for Research in Neuroscience, The Research Institute of the McGill University Health Center, Quebec, Canada
| |
Collapse
|
23
|
Perea JR, Llorens-Martín M, Ávila J, Bolós M. The Role of Microglia in the Spread of Tau: Relevance for Tauopathies. Front Cell Neurosci 2018; 12:172. [PMID: 30042659 PMCID: PMC6048186 DOI: 10.3389/fncel.2018.00172] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 06/01/2018] [Indexed: 01/01/2023] Open
Abstract
Tauopathies are neurodegenerative diseases which course with the accumulation of Tau, mainly in neurons. In addition, Tau accumulates in a hyperphosphorylated and aggregated form. This protein is released into the extracellular space and spreads following a stereotypical pattern, inducing the development of the disease through connected regions of the brain. Microglia-the macrophages of the brain-are involved in maintaining brain homeostasis. They perform a variety of functions related to the surveillance and clearance of pathological proteins, among other dead cells and debris, from the extracellular space that could compromise brain equilibrium. This review focuses on the role played by microglia in tauopathies, specifically in Alzheimer's disease (AD), and how the uncoupling of activation/phagocytosis functions can have fatal consequences leading to the development of the pathology.
Collapse
Affiliation(s)
- Juan R Perea
- Department of Molecular Neuropathology, Centro de Biología Molecular "Severo Ochoa", CBMSO, CSIC, Madrid, Spain.,Network Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - María Llorens-Martín
- Department of Molecular Neuropathology, Centro de Biología Molecular "Severo Ochoa", CBMSO, CSIC, Madrid, Spain.,Department of Molecular Biology, Faculty of Sciences, Universidad Autónoma de Madrid, Madrid, Spain
| | - Jesús Ávila
- Department of Molecular Neuropathology, Centro de Biología Molecular "Severo Ochoa", CBMSO, CSIC, Madrid, Spain.,Network Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Marta Bolós
- Department of Molecular Neuropathology, Centro de Biología Molecular "Severo Ochoa", CBMSO, CSIC, Madrid, Spain.,Network Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| |
Collapse
|
24
|
Li P, Jiang H, Wu H, Wu D, Li H, Yu J, Lai J. AH6809 decreases production of inflammatory mediators by PGE 2 - EP2 - cAMP signaling pathway in an experimentally induced pure cerebral concussion in rats. Brain Res 2018; 1698:11-28. [PMID: 29792868 DOI: 10.1016/j.brainres.2018.05.030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 05/19/2018] [Accepted: 05/20/2018] [Indexed: 12/31/2022]
Abstract
Increasing evidence suggests that PGE2 metabolic pathway is involved in pathological changes of the secondary brain injury after traumatic brain injury. However, the underlying mechanisms, in particular, the correlation between various key enzymes and the brain injury, has remained to be fully explored. More specifically, it remains to be ascertained whether AH6809 (an EP2 receptor antagonist) would interfere with the downstream of the PGE2, regulate the inflammatory mediators and improve neuronal damage in the hippocampus by PGE2 - EP2 - cAMP signaling pathway. The expression and pathological changes of cyclooxygenase-1 (COX-1), cyclooxygenase-2 (COX-2), microsomal prostaglandin-E synthase-1 (mPGES-1), E-prostanoid receptor 2 (EP2), tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β) and inducible nitricoxide synthase (iNOS) in the CA1 area of hippocampus were evaluated by immunohistochemistry, Western blot and RT-PCR after pure cerebral concussion (PCC) induced by a metal pendulum closed brain injury in rats followed by AH6809 treatment. The morphology and number of neurons in CA1 region were analyzed by cresyl violet staining. The concentration of prostaglandin E2 (PGE2) and cyclic adenosine monophosphate (cAMP) was assayed by ELISA. Many neurons in hippocampal CA1 area appeared to undergo necrosis and the number of neurons was concomitantly reduced after PCC injury. With the passage of time, the protein and mRNA expression of various key enzymes including COX-1, COX-2 and mPGES-1, EP2 receptor, and inflammatory mediators including TNF-α, IL-1β and iNOS was increased; meanwhile, the concentration of PGE2 and cAMP was enhanced. After PCC injury given AH6809 intervention, injury of neurons in hippocampal CA1 area was attenuated. The protein and mRNA expression of COX-1, COX-2, mPGES-1, EP2, TNF-α, IL-1β and iNOS was decreased, this was coupled with reduction of PGE2 and cAMP. The results suggest that PGE2 metabolic pathway is involved in secondary pathological changes of PCC. AH6809 improves the recovery of injured neurons in the hippocampal CA1 area and downregulates the inflammatory mediators by PGE2 - EP2 - cAMP signaling pathway.
Collapse
Affiliation(s)
- Ping Li
- College of Forensic Science, Xi'an Jiaotong University, Key Laboratory of Ministry of Public Health for Forensic Science, Xi'an 710061, Shaanxi, PR China; Department of Anatomy and Histology/Embryology, Faculty of Basic Medical Sciences, Kunming Medical University, 1168 West Chunrong Road, Yuhua Avenue, Chenggong District, Kunming 650500, Yunnan, PR China
| | - Hongyan Jiang
- Department of Anatomy and Histology/Embryology, Faculty of Basic Medical Sciences, Kunming Medical University, 1168 West Chunrong Road, Yuhua Avenue, Chenggong District, Kunming 650500, Yunnan, PR China
| | - Haiying Wu
- Department of Emergency and Intensive Care Unit, The First Affiliated Hospital, Kunming Medical University, Kunming 650032, Yunnan, PR China
| | - Deye Wu
- Department of Human Anatomy and Histology/Embryology, Qilu Medical University, 246 West Outer Ring Road, Boshan Economic and Technological Development Zone, Zibo 255213, Shandong, PR China
| | - Hengxi Li
- Department of Anatomy and Histology/Embryology, Faculty of Basic Medical Sciences, Kunming Medical University, 1168 West Chunrong Road, Yuhua Avenue, Chenggong District, Kunming 650500, Yunnan, PR China
| | - Jianyun Yu
- College of Forensic Science and Key Laboratory of Brain Injury, Kunming Medical University, 1168 West Chunrong Road, Yuhua Avenue, Chenggong District, Kunming 650500, Yunnan, PR China
| | - Jianghua Lai
- College of Forensic Science, Xi'an Jiaotong University, Key Laboratory of Ministry of Public Health for Forensic Science, Xi'an 710061, Shaanxi, PR China.
| |
Collapse
|
25
|
He GL, Luo Z, Shen TT, Li P, Yang J, Luo X, Chen CH, Gao P, Yang XS. Inhibition of STAT3- and MAPK-dependent PGE 2 synthesis ameliorates phagocytosis of fibrillar β-amyloid peptide (1-42) via EP2 receptor in EMF-stimulated N9 microglial cells. J Neuroinflammation 2016; 13:296. [PMID: 27871289 PMCID: PMC5117690 DOI: 10.1186/s12974-016-0762-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 11/09/2016] [Indexed: 12/16/2022] Open
Abstract
Background Prostaglandin E2 (PGE2)-involved neuroinflammatory processes are prevalent in several neurological conditions and diseases. Amyloid burden is correlated with the activation of E-prostanoid (EP) 2 receptors by PGE2 in Alzheimer’s disease. We previously demonstrated that electromagnetic field (EMF) exposure can induce pro-inflammatory responses and the depression of phagocytosis in microglial cells, but the signaling pathways involved in phagocytosis of fibrillar β-amyloid (fAβ) in microglial cells exposed to EMF are poorly understood. Given the important role of PGE2 in neural physiopathological processes, we investigated the PGE2-related signaling mechanism in the immunomodulatory phagocytosis of EMF-stimulated N9 microglial cells (N9 cells). Methods N9 cells were exposed to EMF with or without pretreatment with the selective inhibitors of cyclooxygenase-2 (COX-2), Janus kinase 2 (JAK2), signal transducer and activator of transcription 3 (STAT3), and mitogen-activated protein kinases (MAPKs) and antagonists of PG receptors EP1-4. The production of endogenous PGE2 was quantified by enzyme immunoassays. The phagocytic ability of N9 cells was evaluated based on the fluorescence intensity of the engulfed fluorescent-labeled fibrillar β-amyloid peptide (1-42) (fAβ42) measured using a flow cytometer and a fluorescence microscope. The effects of pharmacological agents on EMF-activated microglia were investigated based on the expressions of JAK2, STAT3, p38/ERK/JNK MAPKs, COX-2, microsomal prostaglandin E synthase-1 (mPGES-1), and EP2 using real-time PCR and/or western blotting. Results EMF exposure significantly increased the production of PGE2 and decreased the phagocytosis of fluorescent-labeled fAβ42 by N9 cells. The selective inhibitors of COX-2, JAK2, STAT3, and MAPKs clearly depressed PGE2 release and ameliorated microglial phagocytosis after EMF exposure. Pharmacological agents suppressed the phosphorylation of JAK2-STAT3 and MAPKs, leading to the amelioration of the phagocytic ability of EMF-stimulated N9 cells. Antagonist studies of EP1-4 receptors showed that EMF depressed the phagocytosis of fAβ42 through the PGE2 system, which is linked to EP2 receptors. Conclusions This study indicates that EMF exposure could induce phagocytic depression via JAK2-STAT3- and MAPK-dependent PGE2-EP2 receptor signaling pathways in microglia. Therefore, pharmacological inhibition of PGE2 synthesis and EP2 receptors may be a potential therapeutic strategy to combat the neurobiological deterioration that follows EMF exposure. Electronic supplementary material The online version of this article (doi:10.1186/s12974-016-0762-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Gen-Lin He
- Department of Tropic Hygiene, Institute of Tropical Medicine, Third Military Medical University, 30 Gaotanyan Street, Chongqing, 400038, People's Republic of China
| | - Zhen Luo
- Department of Tropic Hygiene, Institute of Tropical Medicine, Third Military Medical University, 30 Gaotanyan Street, Chongqing, 400038, People's Republic of China
| | - Ting-Ting Shen
- Department of Tropic Hygiene, Institute of Tropical Medicine, Third Military Medical University, 30 Gaotanyan Street, Chongqing, 400038, People's Republic of China
| | - Ping Li
- Department of Tropic Hygiene, Institute of Tropical Medicine, Third Military Medical University, 30 Gaotanyan Street, Chongqing, 400038, People's Republic of China
| | - Ju Yang
- Department of Tropic Hygiene, Institute of Tropical Medicine, Third Military Medical University, 30 Gaotanyan Street, Chongqing, 400038, People's Republic of China
| | - Xue Luo
- Department of Tropic Hygiene, Institute of Tropical Medicine, Third Military Medical University, 30 Gaotanyan Street, Chongqing, 400038, People's Republic of China
| | - Chun-Hai Chen
- Key Laboratory of Medical Protection for Electromagnetic Radiation Ministry of Education, Third Military Medical University, Chongqing, 400038, People's Republic of China
| | - Peng Gao
- Key Laboratory of Medical Protection for Electromagnetic Radiation Ministry of Education, Third Military Medical University, Chongqing, 400038, People's Republic of China
| | - Xue-Sen Yang
- Department of Tropic Hygiene, Institute of Tropical Medicine, Third Military Medical University, 30 Gaotanyan Street, Chongqing, 400038, People's Republic of China.
| |
Collapse
|
26
|
Andreasson KI, Bachstetter AD, Colonna M, Ginhoux F, Holmes C, Lamb B, Landreth G, Lee DC, Low D, Lynch MA, Monsonego A, O’Banion MK, Pekny M, Puschmann T, Russek-Blum N, Sandusky LA, Selenica MLB, Takata K, Teeling J, Town T, Van Eldik LJ, Russek-Blum N, Monsonego A, Low D, Takata K, Ginhoux F, Town T, O’Banion MK, Lamb B, Colonna M, Landreth G, Andreasson KI, Sandusky LA, Selenica MLB, Lee DC, Holmes C, Teeling J, Lynch MA, Van Eldik LJ, Bachstetter AD, Pekny M, Puschmann T. Targeting innate immunity for neurodegenerative disorders of the central nervous system. J Neurochem 2016; 138:653-93. [PMID: 27248001 PMCID: PMC5433264 DOI: 10.1111/jnc.13667] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 04/01/2016] [Accepted: 04/30/2016] [Indexed: 12/21/2022]
Abstract
Neuroinflammation is critically involved in numerous neurodegenerative diseases, and key signaling steps of innate immune activation hence represent promising therapeutic targets. This mini review series originated from the 4th Venusberg Meeting on Neuroinflammation held in Bonn, Germany, 7-9th May 2015, presenting updates on innate immunity in acute brain injury and chronic neurodegenerative disorders, such as traumatic brain injury and Alzheimer disease, on the role of astrocytes and microglia, as well as technical developments that may help elucidate neuroinflammatory mechanisms and establish clinical relevance. In this meeting report, a brief overview of physiological and pathological microglia morphology is followed by a synopsis on PGE2 receptors, insights into the role of arginine metabolism and further relevant aspects of neuroinflammation in various clinical settings, and concluded by a presentation of technical challenges and solutions when working with microglia and astrocyte cultures. Microglial ontogeny and induced pluripotent stem cell-derived microglia, advances of TREM2 signaling, and the cytokine paradox in Alzheimer's disease are further contributions to this article. Neuroinflammation is critically involved in numerous neurodegenerative diseases, and key signaling steps of innate immune activation hence represent promising therapeutic targets. This mini review series originated from the 4th Venusberg Meeting on Neuroinflammation held in Bonn, Germany, 7-9th May 2015, presenting updates on innate immunity in acute brain injury and chronic neurodegenerative disorders, such as traumatic brain injury and Alzheimer's disease, on the role of astrocytes and microglia, as well as technical developments that may help elucidate neuroinflammatory mechanisms and establish clinical relevance. In this meeting report, a brief overview on physiological and pathological microglia morphology is followed by a synopsis on PGE2 receptors, insights into the role of arginine metabolism and further relevant aspects of neuroinflammation in various clinical settings, and concluded by a presentation of technical challenges and solutions when working with microglia cultures. Microglial ontogeny and induced pluripotent stem cell-derived microglia, advances of TREM2 signaling, and the cytokine paradox in Alzheimer's disease are further contributions to this article.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Niva Russek-Blum
- The Dead Sea and Arava Science Center, Central Arava Branch, Yair Station, Hazeva, Israel
| | - Alon Monsonego
- The Shraga Segal Dept. of Microbiology, Immunology and Genetics, The Faculty of Health Sciences: The National Institute of Biotechnology in the Negev, and Zlotowski Center for Neuroscience, Ben-Gurion University, Beer-Sheva 84105, Israel
| | - Donovan Low
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Kazuyuki Takata
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore
- Department of Clinical and Translational Physiology, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Florent Ginhoux
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Terrence Town
- Departments of Physiology and Biophysics, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089,
| | - M. Kerry O’Banion
- Departments of Neuroscience and Neurology, Del Monte Neuromedicine Institute, University of Rochester School of Medicine & Dentistry, Rochester, NY 14642,
| | - Bruce Lamb
- Department of Neurosciences, Cleveland Clinic, Cleveland, OH 44106
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Gary Landreth
- Department of Neurosciences, Case Western Reserve University 44106
| | - Katrin I. Andreasson
- Department of Neurology and Neurological Sciences, Stanford Neuroscience Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Leslie A. Sandusky
- USF Health Byrd Alzheimer’s Institute, Tampa, FL 33613
- College of Pharmacy & Pharmaceutical Sciences, Tampa, FL 33613
| | - Maj-Linda B. Selenica
- USF Health Byrd Alzheimer’s Institute, Tampa, FL 33613
- College of Pharmacy & Pharmaceutical Sciences, Tampa, FL 33613
| | - Daniel C. Lee
- USF Health Byrd Alzheimer’s Institute, Tampa, FL 33613
- College of Pharmacy & Pharmaceutical Sciences, Tampa, FL 33613
| | - Clive Holmes
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Tremona Road, Southampton, SO16 7YD, United Kingdom
| | - Jessica Teeling
- Centre for Biological Sciences, University of Southampton, Southampton General Hospital, Tremona Road, Southampton, SO16 7YD, United Kingdom
| | | | | | | | - Milos Pekny
- Center for Brain Repair and Rehabilitation, Department of Clinical Neuroscience and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, SE-405 30 Gothenburg, Sweden
- Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia
- Hunter Medical Research Institute, University of Newcastle, New South Wales, Australia
| | - Till Puschmann
- Center for Brain Repair and Rehabilitation, Department of Clinical Neuroscience and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, SE-405 30 Gothenburg, Sweden
| |
Collapse
|
27
|
Woodling NS, Andreasson KI. Untangling the Web: Toxic and Protective Effects of Neuroinflammation and PGE2 Signaling in Alzheimer's Disease. ACS Chem Neurosci 2016; 7:454-63. [PMID: 26979823 DOI: 10.1021/acschemneuro.6b00016] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
The neuroinflammatory response has received increasing attention as a key factor in the pathogenesis of Alzheimer's disease (AD). Microglia, the innate immune cells and resident phagocytes of the brain, respond to accumulating Aβ peptides by generating a nonresolving inflammatory response. While this response can clear Aβ peptides from the nervous system in some settings, its failure to do so in AD accelerates synaptic injury, neuronal loss, and cognitive decline. The complex molecular components of this response are beginning to be unraveled, with identification of both damaging and protective roles for individual components of the neuroinflammatory response. Even within one molecular pathway, contrasting effects are often present. As one example, recent studies of the inflammatory cyclooxygenase-prostaglandin pathway have revealed both beneficial and detrimental effects dependent on the disease context, cell type, and downstream signaling pathway. Nonsteroidal anti-inflammatory drugs (NSAIDs), which inhibit cyclooxygenases, are associated with reduced AD risk when taken by cognitively normal populations, but additional clinical and mouse model studies have added complexities and caveats to this finding. Downstream of cyclooxygenase activity, prostaglandin E2 signaling exerts both damaging pro-inflammatory and protective anti-inflammatory effects through actions of specific E-prostanoid G-protein coupled receptors on specific cell types. These complexities underscore the need for careful study of individual components of the neuroinflammatory response to better understand their contribution to AD pathogenesis and progression.
Collapse
Affiliation(s)
- Nathaniel S. Woodling
- Department of Neurology and
Neurological Sciences, Stanford University School of Medicine, 1201
Welch Road, Stanford, California 94305, United States
| | - Katrin I. Andreasson
- Department of Neurology and
Neurological Sciences, Stanford University School of Medicine, 1201
Welch Road, Stanford, California 94305, United States
| |
Collapse
|
28
|
Anastasio TJ. Temporal-logic analysis of microglial phenotypic conversion with exposure to amyloid-β. MOLECULAR BIOSYSTEMS 2016; 11:434-53. [PMID: 25406664 DOI: 10.1039/c4mb00457d] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Alzheimer Disease (AD) remains a leading killer with no adequate treatment. Ongoing research increasingly implicates the brain's immune system as a critical contributor to AD pathogenesis, but the complexity of the immune contribution poses a barrier to understanding. Here I use temporal logic to analyze a computational specification of the immune component of AD. Temporal logic is an extension of logic to propositions expressed in terms of time. It has traditionally been used to analyze computational specifications of complex engineered systems but applications to complex biological systems are now appearing. The inflammatory component of AD involves the responses of microglia to the peptide amyloid-β (Aβ), which is an inflammatory stimulus and a likely causative AD agent. Temporal-logic analysis of the model provides explanations for the puzzling findings that Aβ induces an anti-inflammatory and well as a pro-inflammatory response, and that Aβ is phagocytized by microglia in young but not in old animals. To potentially explain the first puzzle, the model suggests that interferon-γ acts as an "autocrine bridge" over which an Aβ-induced increase in pro-inflammatory cytokines leads to an increase in anti-inflammatory mediators also. To potentially explain the second puzzle, the model identifies a potential instability in signaling via insulin-like growth factor 1 that could explain the failure of old microglia to phagocytize Aβ. The model predicts that augmentation of insulin-like growth factor 1 signaling, and activation of protein kinase C in particular, could move old microglia from a neurotoxic back toward a more neuroprotective and phagocytic phenotype.
Collapse
Affiliation(s)
- Thomas J Anastasio
- Computational Neurobiology Laboratory, Department of Molecular and Integrative Physiology, and Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
| |
Collapse
|
29
|
Curcumin Ameliorates the Reduction Effect of PGE2 on Fibrillar β-Amyloid Peptide (1-42)-Induced Microglial Phagocytosis through the Inhibition of EP2-PKA Signaling in N9 Microglial Cells. PLoS One 2016; 11:e0147721. [PMID: 26824354 PMCID: PMC4732694 DOI: 10.1371/journal.pone.0147721] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 01/07/2016] [Indexed: 01/18/2023] Open
Abstract
Inflammatory activation of microglia and β amyloid (Aβ) deposition are considered to work both independently and synergistically to contribute to the increased risk of Alzheimer's disease (AD). Recent studies indicate that long-term use of phenolic compounds provides protection against AD, primarily due to their anti-inflammatory actions. We previously suggested that phenolic compound curcumin ameliorated phagocytosis possibly through its anti-inflammatory effects rather than direct regulation of phagocytic function in electromagnetic field-exposed N9 microglial cells (N9 cells). Here, we explored the prostaglandin-E2 (PGE2)-related signaling pathway that involved in curcumin-mediated phagocytosis in fibrillar β-amyloid peptide (1-42) (fAβ42)-stimulated N9 cells. Treatment with fAβ42 increased phagocytosis of fluorescent-labeled latex beads in N9 cells. This increase was attenuated in a dose-dependent manner by endogenous and exogenous PGE2, as well as a selective EP2 or protein kinase A (PKA) agonist, but not by an EP4 agonist. We also found that an antagonist of EP2, but not EP4, abolished the reduction effect of PGE2 on fAβ42-induced microglial phagocytosis. Additionally, the increased expression of endogenous PGE2, EP2, and cyclic adenosine monophosphate (AMP), and activation of vasodilator-stimulated phosphoprotein, cyclic AMP responsive element-binding protein, and PKA were depressed by curcumin administration. This reduction led to the amelioration of the phagocytic abilities of PGE2-stimulated N9 cells. Taken together, these data suggested that curcumin restored the attenuating effect of PGE2 on fAβ42-induced microglial phagocytosis via a signaling mechanism involving EP2 and PKA. Moreover, due to its immune modulatory effects, curcumin may be a promising pharmacological candidate for neurodegenerative diseases.
Collapse
|
30
|
von Bernhardi R, Heredia F, Salgado N, Muñoz P. Microglia Function in the Normal Brain. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 949:67-92. [PMID: 27714685 DOI: 10.1007/978-3-319-40764-7_4] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The activation of microglia has been recognized for over a century by their morphological changes. Long slender microglia acquire a short sturdy ramified shape when activated. During the past 20 years, microglia have been accepted as an essential cellular component for understanding the pathogenic mechanism of many brain diseases, including neurodegenerative diseases. More recently, functional studies and imaging in mouse models indicate that microglia are active in the healthy central nervous system. It has become evident that microglia release several signal molecules that play key roles in the crosstalk among brain cells, i.e., astrocytes and oligodendrocytes with neurons, as well as with regulatory immune cells. Recent studies also reveal the heterogeneous nature of microglia diverse functions depending on development, previous exposure to stimulation events, brain region of residence, or pathological state. Subjects to approach by future research are still the unresolved questions regarding the conditions and mechanisms that render microglia protective, capable of preventing or reducing damage, or deleterious, capable of inducing or facilitating the progression of neuropathological diseases. This novel knowledge will certainly change our view on microglia as therapeutic target, shifting our goal from their general silencing to the generation of treatments able to change their activation pattern.
Collapse
Affiliation(s)
- Rommy von Bernhardi
- Escuela de Medicina. Departamento de Neurología, Pontificia Universidad Católica de Chile, Marcoleta 391, Santiago, Chile.
| | - Florencia Heredia
- Escuela de Medicina. Departamento de Neurología, Pontificia Universidad Católica de Chile, Marcoleta 391, Santiago, Chile
| | - Nicole Salgado
- Escuela de Medicina. Departamento de Neurología, Pontificia Universidad Católica de Chile, Marcoleta 391, Santiago, Chile
| | - Paola Muñoz
- Escuela de Medicina. Departamento de Neurología, Pontificia Universidad Católica de Chile, Marcoleta 391, Santiago, Chile
| |
Collapse
|
31
|
Pathophysiological Roles of Cyclooxygenases and Prostaglandins in the Central Nervous System. Mol Neurobiol 2015; 53:4754-71. [PMID: 26328537 DOI: 10.1007/s12035-015-9355-3] [Citation(s) in RCA: 135] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Accepted: 07/07/2015] [Indexed: 01/01/2023]
Abstract
Cyclooxygenases (COXs) oxidize arachidonic acid to prostaglandin (PG) G2 and H2 followed by PG synthases that generates PGs and thromboxane (TX) A2. COXs are divided into COX-1 and COX-2. In the central nervous system, COX-1 is constitutively expressed in neurons, astrocytes, and microglial cells. COX-2 is upregulated in these cells under pathophysiological conditions. In hippocampal long-term potentiation, COX-2, PGE synthase, and PGE2 are induced in post-synaptic neurons. PGE2 acts pre-synaptic EP2 receptor, generates cAMP, stimulates protein kinase A, modulates voltage-dependent calcium channel, facilitates glutamatergic synaptic transmission, and potentiates long-term plasticity. PGD2, PGE2, and PGI2 exhibit neuroprotective effects via Gs-coupled DP1, EP2/EP4, and IP receptors, respectively. COX-2, PGD2, PGE2, PGF2α, and TXA2 are elevated in stroke. COX-2 inhibitors exhibit neuroprotective effects in vivo and in vitro models of stroke, Alzheimer's disease, Parkinson's disease, multiple sclerosis, amyotrophic lateral sclerosis, epilepsy, and schizophrenia, suggesting neurotoxicities of COX products. PGE2, PGF2α, and TXA2 can contribute to the neurodegeneration via EP1, FP, and TP receptors, respectively, which are coupled with Gq, stimulate phospholipase C and cleave phosphatidylinositol diphosphate to produce inositol triphosphate and diacylglycerol. Inositol triphosphate binds to inositol triphosphate receptor in endoplasmic reticulum, releases calcium, and results in increasing intracellular calcium concentrations. Diacylglycerol activates calcium-dependent protein kinases. PGE2 disrupts Ca(2+) homeostasis by impairing Na(+)-Ca(2+) exchange via EP1, resulting in the excess Ca(2+) accumulation. Neither PGE2, PGF2α, nor TXA2 causes neuronal cell death by itself, suggesting that they might enhance the ischemia-induced neurodegeneration. Alternatively, PGE2 is non-enzymatically dehydrated to a cyclopentenone PGA2, which induces neuronal cell death. Although PGD2 induces neuronal apoptosis after a lag time, neither DP1 nor DP2 is involved in the neurotoxicity. As well as PGE2, PGD2 is non-enzymatically dehydrated to a cyclopentenone 15-deoxy-Δ(12,14)-PGJ2, which induces neuronal apoptosis without a lag time. However, neurotoxicities of these cyclopentenones are independent of their receptors. The COX-2 inhibitor inhibits both the anchorage-dependent and anchorage-independent growth of glioma cell lines regardless of COX-2 expression, suggesting that some COX-2-independent mechanisms underlie the antineoplastic effect of the inhibitor. PGE2 attenuates this antineoplastic effect, suggesting that the predominant mechanism is COX-dependent. COX-2 or EP1 inhibitors show anti-neoplastic effects. Thus, our review presents evidences for pathophysiological roles of cyclooxygenases and prostaglandins in the central nervous system.
Collapse
|
32
|
Johansson JU, Woodling NS, Brown HD, Wang Q, Andreasson KI. Microarray analysis of the in vivo response of microglia to Aβ peptides in mice with conditional deletion of the prostaglandin EP2 receptor. GENOMICS DATA 2015; 5:268-271. [PMID: 26251825 PMCID: PMC4522932 DOI: 10.1016/j.gdata.2015.06.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Amyloid-β (Aβ) peptides accumulate in the brains of patients with Alzheimer's disease (AD), where they generate a persistent inflammatory response from microglia, the innate immune cells of the brain. The immune modulatory cyclooxygenase/prostaglandin E2 (COX/PGE2) pathway has been implicated in preclinical AD development, both in human epidemiology studies and in transgenic rodent models of AD [2,3]. PGE2 signals through four G-protein-coupled receptors, including the EP2 receptor that has been investigated for its role in mediating the inflammatory and phagocytic responses to Aβ [4]. To identify transcriptional differences in microglia lacking the EP2 receptor, we examined mice with EP2 conditionally deleted in Cd11b-expressing immune cells. We injected Aβ peptides or saline vehicle into the brains of adult mice, isolated primary microglia, and analyzed RNA expression by microarray. The resulting datasets were analyzed in two studies [5,6], one describing the basal status of microglia with or without EP2 deletion, and the second study analyzing the microglial response to Aβ. Here we describe in detail the experimental design and data analyses. The raw data from these studies are deposited in GEO, accession GSE57181 (http://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE57181).
Collapse
Affiliation(s)
- Jenny U Johansson
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305
| | - Nathaniel S Woodling
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305
| | - Holden D Brown
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305
| | - Qian Wang
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305
| | - Katrin I Andreasson
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305
| |
Collapse
|
33
|
Johansson JU, Woodling NS, Shi J, Andreasson KI. Inflammatory Cyclooxygenase Activity and PGE 2 Signaling in Models of Alzheimer's Disease. ACTA ACUST UNITED AC 2015; 11:125-131. [PMID: 28413375 PMCID: PMC5384338 DOI: 10.2174/1573395511666150707181414] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Revised: 02/03/2015] [Accepted: 04/19/2015] [Indexed: 11/28/2022]
Abstract
The inflammatory response is a fundamental driving force in the pathogenesis of Alzheimer’s disease (AD). In the setting of accumulating immunogenic Aß peptide assemblies, microglia, the innate immune cells of the brain, generate a non-resolving immune response and fail to adequately clear accumulating Aß peptides, accelerating neuronal and synaptic injury. Pathological, biomarker, and imaging studies point to a prominent role of the innate immune response in AD development, and the molecular components of this response are beginning to be unraveled. The inflammatory cyclooxygenase-PGE2 pathway is implicated in pre-clinical development of AD, both in epidemiology of normal aging populations and in transgenic mouse models of Familial AD. The cyclooxygenase-PGE2 pathway modulates the inflammatory response to accumulating Aß peptides through actions of specific E-prostanoid G-protein coupled receptors.
Collapse
Affiliation(s)
- Jenny U Johansson
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA.,Present address: SRI International, Menlo Park, CA, USA
| | - Nathaniel S Woodling
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA.,Present address: Institute of Healthy Ageing, University College London, London, UK
| | - Ju Shi
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA.,Present address: True North Therapeutics, South San Francisco, CA, USA
| | - Katrin I Andreasson
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
34
|
Fox BM, Beck HP, Roveto PM, Kayser F, Cheng Q, Dou H, Williamson T, Treanor J, Liu H, Jin L, Xu G, Ma J, Wang S, Olson SH. A Selective Prostaglandin E2 Receptor Subtype 2 (EP2) Antagonist Increases the Macrophage-Mediated Clearance of Amyloid-Beta Plaques. J Med Chem 2015; 58:5256-73. [DOI: 10.1021/acs.jmedchem.5b00567] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Brian M. Fox
- Amgen South San Francisco, Amgen Inc., 1120 Veterans
Boulevard, South San Francisco, California 94080, United States
| | - Hilary P. Beck
- Amgen South San Francisco, Amgen Inc., 1120 Veterans
Boulevard, South San Francisco, California 94080, United States
| | - Philip M. Roveto
- Amgen South San Francisco, Amgen Inc., 1120 Veterans
Boulevard, South San Francisco, California 94080, United States
| | - Frank Kayser
- Amgen South San Francisco, Amgen Inc., 1120 Veterans
Boulevard, South San Francisco, California 94080, United States
| | - Qingwen Cheng
- Amgen South San Francisco, Amgen Inc., 1120 Veterans
Boulevard, South San Francisco, California 94080, United States
| | - Hannah Dou
- Amgen South San Francisco, Amgen Inc., 1120 Veterans
Boulevard, South San Francisco, California 94080, United States
| | - Toni Williamson
- Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - James Treanor
- Amgen South San Francisco, Amgen Inc., 1120 Veterans
Boulevard, South San Francisco, California 94080, United States
| | - Hantao Liu
- Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Lixia Jin
- Amgen South San Francisco, Amgen Inc., 1120 Veterans
Boulevard, South San Francisco, California 94080, United States
| | - Guifen Xu
- Amgen South San Francisco, Amgen Inc., 1120 Veterans
Boulevard, South San Francisco, California 94080, United States
| | - Ji Ma
- Amgen South San Francisco, Amgen Inc., 1120 Veterans
Boulevard, South San Francisco, California 94080, United States
| | - Songli Wang
- Amgen South San Francisco, Amgen Inc., 1120 Veterans
Boulevard, South San Francisco, California 94080, United States
| | - Steven H. Olson
- Amgen South San Francisco, Amgen Inc., 1120 Veterans
Boulevard, South San Francisco, California 94080, United States
| |
Collapse
|
35
|
Leclerc JL, Lampert AS, Diller MA, Immergluck JB, Doré S. Prostaglandin E2 EP2 receptor deletion attenuates intracerebral hemorrhage-induced brain injury and improves functional recovery. ASN Neuro 2015; 7:7/2/1759091415578713. [PMID: 25873308 PMCID: PMC4720177 DOI: 10.1177/1759091415578713] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Intracerebral hemorrhage (ICH) is a devastating type of stroke characterized by bleeding into the brain parenchyma and secondary brain injury resulting from strong neuroinflammatory responses to blood components. Production of prostaglandin E2 (PGE2) is significantly upregulated following ICH and contributes to this inflammatory response in part through its E prostanoid receptor subtype 2 (EP2). Signaling through the EP2 receptor has been shown to affect outcomes of many acute and chronic neurological disorders; although, not yet explored in the context of ICH. Wildtype (WT) and EP2 receptor knockout (EP2−/−) mice were subjected to ICH, and various anatomical and functional outcomes were assessed by histology and neurobehavioral testing, respectively. When compared with age-matched WT controls, EP2−/− mice had 41.9 ± 4.7% smaller ICH-induced brain lesions and displayed significantly less ipsilateral hemispheric enlargement and incidence of intraventricular hemorrhage. Anatomical outcomes correlated with improved functional recovery as identified by neurological deficit scoring. Histological staining was performed to begin investigating the mechanisms involved in EP2-mediated neurotoxicity after ICH. EP2−/− mice exhibited 45.5 ± 5.8% and 41.4 ± 8.1% less blood and ferric iron accumulation, respectively. Furthermore, significantly less striatal and cortical microgliosis, striatal and cortical astrogliosis, blood–brain barrier breakdown, and peripheral neutrophil infiltration were seen in EP2−/− mice. This study is the first to suggest a deleterious role for the PGE2-EP2 signaling axis in modulating brain injury, inflammation, and functional recovery following ICH. Targeting the EP2 G protein-coupled receptor may represent a new therapeutic avenue for the treatment of hemorrhagic stroke.
Collapse
Affiliation(s)
- Jenna L Leclerc
- Department of Anesthesiology, University of Florida, Gainesville, FL, USA Department of Neuroscience, University of Florida, Gainesville, FL, USA
| | - Andrew S Lampert
- Department of Anesthesiology, University of Florida, Gainesville, FL, USA
| | - Matthew A Diller
- Department of Anesthesiology, University of Florida, Gainesville, FL, USA
| | | | - Sylvain Doré
- Department of Anesthesiology, University of Florida, Gainesville, FL, USA Department of Neuroscience, University of Florida, Gainesville, FL, USA Departments of Neurology, Psychiatry, and Pharmaceutics, University of Florida, Gainesville, FL, USA
| |
Collapse
|
36
|
Ganesh T. Evaluation of WO 2012/177618 A1 and US-2014/0179750 A1: novel small molecule antagonists of prostaglandin-E2 receptor EP2. Expert Opin Ther Pat 2015; 25:837-44. [PMID: 25772215 DOI: 10.1517/13543776.2015.1025752] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Recent studies underscore that prostaglandin-E2 exerts mostly proinflammatory effects in chronic CNS and peripheral disease models, mainly through a specific prostanoid receptor EP2. However, very few highly characterized EP2 receptor antagonists have been reported until recently, when Pfizer and Emory University published two distinct classes of EP2 antagonists with good potency, selectivity and pharmacokinetics. The purpose of this article is to evaluate recently published patents WO 2012/177618 A1 and US-2014/0179750 A1 from Emory, which describe a number of cinnamic amide- and amide-derivatives as a potent antagonists of EP2 receptor, and their neuroprotective effects in in vitro and in an in vivo model. A selected compound from this patent(s) also attenuates prostate cancer cell growth and invasion in vitro, suggesting these compounds should be developed for therapeutic use.
Collapse
Affiliation(s)
- Thota Ganesh
- Emory University School of Medicine, Department of Pharmacology , 1510 Clifton Rd, Atlanta, GA 30322 , USA +1 404 727 7393 ; +1 404 727 0365 ;
| |
Collapse
|
37
|
Lee YH, Lin CH, Hsu PC, Sun YY, Huang YJ, Zhuo JH, Wang CY, Gan YL, Hung CC, Kuan CY, Shie FS. Aryl hydrocarbon receptor mediates both proinflammatory and anti-inflammatory effects in lipopolysaccharide-activated microglia. Glia 2015; 63:1138-54. [PMID: 25690886 DOI: 10.1002/glia.22805] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Accepted: 01/21/2015] [Indexed: 12/27/2022]
Abstract
The aryl hydrocarbon receptor (AhR) regulates peripheral immunity; but its role in microglia-mediated neuroinflammation in the brain remains unknown. Here, we demonstrate that AhR mediates both anti-inflammatory and proinflammatory effects in lipopolysaccharide (LPS)-activated microglia. Activation of AhR by its ligands, formylindolo[3,2-b]carbazole (FICZ) or 3-methylcholanthrene (3MC), attenuated LPS-induced microglial immune responses. AhR also showed proinflammatory effects, as evidenced by the findings that genetic silence of AhR ameliorated the LPS-induced microglial immune responses and LPS-activated microglia-mediated neurotoxicity. Similarly, LPS-induced expressions of tumor necrosis factor α (TNFα) and inducible nitric oxide synthase (iNOS) were reduced in the cerebral cortex of AhR-deficient mice. Intriguingly, LPS upregulated and activated AhR in the absence of AhR ligands via the MEK1/2 signaling pathway, which effects were associated with a transient inhibition of cytochrome P450 1A1 (CYP1A1). Although AhR ligands synergistically enhance LPS-induced AhR activation, leading to suppression of LPS-induced microglial immune responses, they cannot do so on their own in microglia. Chromatin immunoprecipitation results further revealed that LPS-FICZ co-treatment, but not LPS alone, not only resulted in co-recruitment of both AhR and NFκB onto the κB site of TNFα gene promoter but also reduced LPS-induced AhR binding to the DRE site of iNOS gene promoter. Together, we provide evidence showing that microglial AhR, which can be activated by LPS, exerts bi-directional effects on the regulation of LPS-induced neuroinflammation, depending on the availability of external AhR ligands. These findings confer further insights into the potential link between environmental factors and the inflammatory brain disorders.
Collapse
Affiliation(s)
- Yi-Hsuan Lee
- Institute of Physiology, National Yang-Ming University, Taipei, Taiwan; Brain Research Center, National Yang-Ming University, Taipei, Taiwan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Jiang J, Yang MS, Quan Y, Gueorguieva P, Ganesh T, Dingledine R. Therapeutic window for cyclooxygenase-2 related anti-inflammatory therapy after status epilepticus. Neurobiol Dis 2015; 76:126-136. [PMID: 25600211 DOI: 10.1016/j.nbd.2014.12.032] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Revised: 12/12/2014] [Accepted: 01/09/2015] [Indexed: 11/17/2022] Open
Abstract
As a prominent inflammatory effector of cyclooxygenase-2 (COX-2), prostaglandin E2 (PGE2) mediates brain inflammation and injury in many chronic central nervous system (CNS) conditions including seizures and epilepsy, largely through its receptor subtype EP2. However, EP2 receptor activation might also be neuroprotective in models of excitotoxicity and ischemia. These seemingly incongruent observations expose the delicacy of immune and inflammatory signaling in the brain; thus the therapeutic window for quelling neuroinflammation might vary with injury type and target molecule. Here, we identify a therapeutic window for EP2 antagonism to reduce delayed mortality and functional morbidity after status epilepticus (SE) in mice. Importantly, treatment must be delayed relative to SE onset to be effective, a finding that could be explained by the time-course of COX-2 induction after SE and compound pharmacokinetics. A large number of inflammatory mediators were upregulated in hippocampus after SE with COX-2 and IL-1β temporally leading many others. Thus, EP2 antagonism represents a novel anti-inflammatory strategy to treat SE with a tightly-regulated therapeutic window.
Collapse
Affiliation(s)
- Jianxiong Jiang
- Department of Pharmacology, School of Medicine, Emory University, Atlanta, GA 30322, United States; Division of Pharmaceutical sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH 45267, United States.
| | - Myung-Soon Yang
- Department of Pharmacology, School of Medicine, Emory University, Atlanta, GA 30322, United States
| | - Yi Quan
- Department of Pharmacology, School of Medicine, Emory University, Atlanta, GA 30322, United States
| | - Paoula Gueorguieva
- Department of Pharmacology, School of Medicine, Emory University, Atlanta, GA 30322, United States
| | - Thota Ganesh
- Department of Pharmacology, School of Medicine, Emory University, Atlanta, GA 30322, United States
| | - Raymond Dingledine
- Department of Pharmacology, School of Medicine, Emory University, Atlanta, GA 30322, United States
| |
Collapse
|
39
|
Krauthausen M, Kummer MP, Zimmermann J, Reyes-Irisarri E, Terwel D, Bulic B, Heneka MT, Müller M. CXCR3 promotes plaque formation and behavioral deficits in an Alzheimer's disease model. J Clin Invest 2014; 125:365-78. [PMID: 25500888 DOI: 10.1172/jci66771] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2012] [Accepted: 11/10/2014] [Indexed: 01/28/2023] Open
Abstract
Chemokines are important modulators of neuroinflammation and neurodegeneration. In the brains of Alzheimer's disease (AD) patients and in AD animal models, the chemokine CXCL10 is found in high concentrations, suggesting a pathogenic role for this chemokine and its receptor, CXCR3. Recent studies aimed at addressing the role of CXCR3 in neurological diseases indicate potent, but diverse, functions for CXCR3. Here, we examined the impact of CXCR3 in the amyloid precursor protein (APP)/presenilin 1 (PS1) transgenic mouse model of AD. We found that, compared with control APP/PSI animals, plaque burden and Aβ levels were strongly reduced in CXCR3-deficient APP/PS1 mice. Analysis of microglial phagocytosis in vitro and in vivo demonstrated that CXCR3 deficiency increased the microglial uptake of Aβ. Application of a CXCR3 antagonist increased microglial Aβ phagocytosis, which was associated with reduced TNF-α secretion. Moreover, in CXCR3-deficient APP/PS1 mice, microglia exhibited morphological activation and reduced plaque association, and brain tissue from APP/PS1 animals lacking CXCR3 had reduced concentrations of proinflammatory cytokines compared with controls. Further, loss of CXCR3 attenuated the behavioral deficits observed in APP/PS1 mice. Together, our data indicate that CXCR3 signaling mediates development of AD-like pathology in APP/PS1 mice and suggest that CXCR3 has potential as a therapeutic target for AD.
Collapse
|
40
|
Li X, Melief E, Postupna N, Montine KS, Keene CD, Montine TJ. Prostaglandin E2 receptor subtype 2 regulation of scavenger receptor CD36 modulates microglial Aβ42 phagocytosis. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 185:230-9. [PMID: 25452117 DOI: 10.1016/j.ajpath.2014.09.016] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2013] [Revised: 09/14/2014] [Accepted: 09/18/2014] [Indexed: 12/18/2022]
Abstract
Recent studies underline the potential relevance of microglial innate immune activation in Alzheimer disease. Primary mouse microglia that lack prostaglandin E2 receptor subtype 2 (EP2) show decreased innate immune-mediated neurotoxicity and increased amyloid β (Aβ) peptide phagocytosis, features that were replicated in vivo. Here, we tested the hypothesis that scavenger receptor CD36 is an effector of EP2-regulated Aβ phagocytosis. CD36 expression was 143-fold greater in mouse primary microglia than in primary astrocytes. Three different means of suppressing EP2 signaling increased and an agonist of EP2 decreased CD36 expression in primary wild-type microglia. Activation of Toll-like receptor (TLR) 3, TLR4, and TLR7, but not TLR2 or TLR9, reduced primary microglial CD36 transcription and cell surface CD36 protein and reduced Aβ42 phagocytosis as well. At each step, the effects of innate immune activation on CD36 were reversed by at least 50% by an EP2 antagonist, and this partial rescue of microglia Aβ42 phagocytosis was largely mediated by CD36 activity. Finally, we showed in hippocampus of wild-type mice that innate immune activation suppressed CD36 expression by an EP2-dependent mechanism. Taken together with results of others that found brain clearance of Aβ peptides and behavioral improvements mediated by CD36 in mice, regulation of CD36-mediated Aβ phagocytosis by suppression of EP2 signaling may provide a new approach to suppressing some aspects of Alzheimer disease pathogenesis.
Collapse
Affiliation(s)
- Xianwu Li
- Department of Pathology, University of Washington, Seattle, Washington.
| | - Erica Melief
- Department of Pathology, University of Washington, Seattle, Washington
| | - Nadia Postupna
- Department of Pathology, University of Washington, Seattle, Washington
| | | | - C Dirk Keene
- Department of Pathology, University of Washington, Seattle, Washington
| | - Thomas J Montine
- Department of Pathology, University of Washington, Seattle, Washington
| |
Collapse
|
41
|
Erb L, Cao C, Ajit D, Weisman GA. P2Y receptors in Alzheimer's disease. Biol Cell 2014; 107:1-21. [PMID: 25179475 DOI: 10.1111/boc.201400043] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Accepted: 08/27/2014] [Indexed: 02/06/2023]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia, affecting more than 10% of people over the age of 65. Age is the greatest risk factor for AD, although a combination of genetic, lifestyle and environmental factors also contribute to disease development. Common features of AD are the formation of plaques composed of beta-amyloid peptides (Aβ) and neuronal death in brain regions involved in learning and memory. Although Aβ is neurotoxic, the primary mechanisms by which Aβ affects AD development remain uncertain and controversial. Mouse models overexpressing amyloid precursor protein and Aβ have revealed that Aβ has potent effects on neuroinflammation and cerebral blood flow that contribute to AD progression. Therefore, it is important to consider how endogenous signalling in the brain responds to Aβ and contributes to AD pathology. In recent years, Aβ has been shown to affect ATP release from brain and blood cells and alter the expression of G protein-coupled P2Y receptors that respond to ATP and other nucleotides. Accumulating evidence reveals a prominent role for P2Y receptors in AD pathology, including Aβ production and elimination, neuroinflammation, neuronal function and cerebral blood flow.
Collapse
Affiliation(s)
- Laurie Erb
- Department of Biochemistry, Life Sciences Center, University of Missouri, Columbia, MO, 65211, U.S.A
| | | | | | | |
Collapse
|
42
|
Suppression of Alzheimer-associated inflammation by microglial prostaglandin-E2 EP4 receptor signaling. J Neurosci 2014; 34:5882-94. [PMID: 24760848 DOI: 10.1523/jneurosci.0410-14.2014] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
A persistent and nonresolving inflammatory response to accumulating Aβ peptide species is a cardinal feature in the development of Alzheimer's disease (AD). In response to accumulating Aβ peptide species, microglia, the innate immune cells of the brain, generate a toxic inflammatory response that accelerates synaptic and neuronal injury. Many proinflammatory signaling pathways are linked to progression of neurodegeneration. However, endogenous anti-inflammatory pathways capable of suppressing Aβ-induced inflammation represent a relatively unexplored area. Here we report that signaling through the prostaglandin-E2 (PGE2) EP4 receptor potently suppresses microglial inflammatory responses to Aβ42 peptides. In cultured microglial cells, EP4 stimulation attenuated levels of Aβ42-induced inflammatory factors and potentiated phagocytosis of Aβ42. Microarray analysis demonstrated that EP4 stimulation broadly opposed Aβ42-driven gene expression changes in microglia, with enrichment for targets of IRF1, IRF7, and NF-κB transcription factors. In vivo, conditional deletion of microglial EP4 in APPSwe-PS1ΔE9 (APP-PS1) mice conversely increased inflammatory gene expression, oxidative protein modification, and Aβ deposition in brain at early stages of pathology, but not at later stages, suggesting an early anti-inflammatory function of microglial EP4 signaling in the APP-PS1 model. Finally, EP4 receptor levels decreased significantly in human cortex with progression from normal to AD states, suggesting that early loss of this beneficial signaling system in preclinical AD development may contribute to subsequent progression of pathology.
Collapse
|
43
|
Ganesh T, Jiang J, Yang MS, Dingledine R. Lead optimization studies of cinnamic amide EP2 antagonists. J Med Chem 2014; 57:4173-84. [PMID: 24773616 PMCID: PMC4032197 DOI: 10.1021/jm5000672] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Prostanoid receptor EP2 can play a proinflammatory role, exacerbating disease pathology in a variety of central nervous system and peripheral diseases. A highly selective EP2 antagonist could be useful as a drug to mitigate the inflammatory consequences of EP2 activation. We recently identified a cinnamic amide class of EP2 antagonists. The lead compound in this class (5d) displays anti-inflammatory and neuroprotective actions. However, this compound exhibited moderate selectivity to EP2 over the DP1 prostanoid receptor (∼10-fold) and low aqueous solubility. We now report compounds that display up to 180-fold selectivity against DP1 and up to 9-fold higher aqueous solubility than our previous lead. The newly developed compounds also display higher selectivity against EP4 and IP receptors and a comparable plasma pharmacokinetics. Thus, these compounds are useful for proof of concept studies in a variety of models where EP2 activation is playing a deleterious role.
Collapse
Affiliation(s)
- Thota Ganesh
- Department of Pharmacology, School of Medicine, Emory University , 1510 Clifton Road, Atlanta, Georgia 30322, United States
| | | | | | | |
Collapse
|
44
|
Singh Bahia M, Kumar Katare Y, Silakari O, Vyas B, Silakari P. Inhibitors of Microsomal Prostaglandin E2
Synthase-1 Enzyme as Emerging Anti-Inflammatory Candidates. Med Res Rev 2014; 34:825-55. [DOI: 10.1002/med.21306] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Malkeet Singh Bahia
- Molecular Modelling Lab (MML); Department of Pharmaceutical Sciences and Drug Research; Punjabi University; Patiala Punjab 147002 India
| | - Yogesh Kumar Katare
- Radharaman Institute of Pharmaceutical Sciences; Bhopal Madhya Pradesh 462046 India
| | - Om Silakari
- Molecular Modelling Lab (MML); Department of Pharmaceutical Sciences and Drug Research; Punjabi University; Patiala Punjab 147002 India
| | - Bhawna Vyas
- Department of Chemistry; Punjabi University; Patiala Punjab 147002 India
| | - Pragati Silakari
- Adina institute of Pharmaceutical Sciences; Sagar Madhya Pradesh (M.P.) 470001 India
| |
Collapse
|
45
|
Abstract
Cycoloxygenase-2 (COX-2) induction is prevalent in a variety of (brain and peripheral) injury models where COX-2 levels correlate with disease progression. Thus, COX-2 has been widely explored for anti-inflammatory therapy with COX-2 inhibitors, which proved to be effective in reducing the pain and inflammation in patients with arthritis and menstrual cramps, but they have not provided any benefit to patients with chronic inflammatory neurodegenerative disease. Recently, two COX-2 drugs, rofecoxib and valdecoxib, were withdrawn from the United States market due to cardiovascular side effects. Thus, future anti-inflammatory therapy could be targeted through a specific prostanoid receptor downstream of COX-2. The PGE2 receptor EP2 is emerging as a pro-inflammatory target in a variety of CNS and peripheral diseases. Here we highlight the latest developments on the role of EP2 in diseases, mechanism of activation, and small molecule discovery targeted either to enhance or to block the function of this receptor.
Collapse
Affiliation(s)
- Thota Ganesh
- Department of Pharmacology, Emory University School of Medicine , 1510 Clifton Road, Atlanta, Georgia, 30322, United States
| |
Collapse
|
46
|
Ajit D, Woods LT, Camden JM, Thebeau CN, El-Sayed FG, Greeson GW, Erb L, Petris MJ, Miller DC, Sun GY, Weisman GA. Loss of P2Y₂ nucleotide receptors enhances early pathology in the TgCRND8 mouse model of Alzheimer's disease. Mol Neurobiol 2013; 49:1031-42. [PMID: 24193664 DOI: 10.1007/s12035-013-8577-5] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Accepted: 10/21/2013] [Indexed: 11/26/2022]
Abstract
Neuroinflammation is a prominent feature in Alzheimer's disease (AD) and activation of the brain's innate immune system, particularly microglia, has been postulated to both retard and accelerate AD progression. Recent studies indicate that the G protein-coupled P2Y2 nucleotide receptor (P2Y2R) is an important regulator of innate immunity by assisting in the recruitment of monocytes to injured tissue, neutrophils to bacterial infections and eosinophils to allergen-infected lungs. In this study, we investigated the role of the P2Y2R in progression of an AD-like phenotype in the TgCRND8 mouse model that expresses Swedish and Indiana mutations in amyloid precursor protein (APP). Our results indicate that P2Y 2 R expression is upregulated in TgCRND8 mouse brain within 10 weeks of age and then decreases after 25 weeks of age, as compared to littermate controls expressing low levels of the P2Y 2 R. TgCRND8 mice with homozygous P2Y 2 R deletion survive less than 5 weeks, whereas mice with heterozygous P2Y 2 R deletion survive for 12 weeks, a time point when TgCRND8 mice are fully viable. Heterozygous P2Y 2 R deletion in TgCRND8 mice increased β-amyloid (Aβ) plaque load and soluble Aβ1-42 levels in the cerebral cortex and hippocampus, decreased the expression of the microglial marker CD11b in these brain regions and caused neurological deficits within 10 weeks of age, as compared to age-matched TgCRND8 mice. These findings suggest that the P2Y2R is important for the recruitment and activation of microglial cells in the TgCRND8 mouse brain and that the P2Y2R may regulate neuroprotective mechanisms through microglia-mediated clearance of Aβ that when lost can accelerate the onset of an AD-like phenotype in the TgCRND8 mouse.
Collapse
Affiliation(s)
- Deepa Ajit
- Department of Biochemistry, University of Missouri, 540E Life Sciences Center, 1201 Rollins Road, Columbia, MO, 65211-7310, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Tsay HJ, Huang YC, Huang FL, Chen CP, Tsai YC, Wang YH, Wu MF, Chiang FY, Shiao YJ. Amyloid β peptide-mediated neurotoxicity is attenuated by the proliferating microglia more potently than by the quiescent phenotype. J Biomed Sci 2013; 20:78. [PMID: 24152138 PMCID: PMC3870991 DOI: 10.1186/1423-0127-20-78] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Accepted: 10/20/2013] [Indexed: 12/12/2022] Open
Abstract
Background The specific role of microglia on Aβ-mediated neurotoxicity is difficult to assign in vivo due to their complicated environment in the brain. Therefore, most of the current microglia-related studies employed the isolated microglia. However, the previous in vitro studies have suggested either beneficial or destructive function in microglia. Therefore, to investigate the phenotypes of the isolated microglia which exert activity of neuroprotective or destructive is required. Results The present study investigates the phenotypes of isolated microglia on protecting neuron against Aβ-mediated neurotoxicity. Primary microglia were isolated from the mixed glia culture, and were further cultured to distinct phenotypes, designated as proliferating amoeboid microglia (PAM) and differentiated process-bearing microglia (DPM). Their inflammatory phenotypes, response to amyloid β (Aβ), and the beneficial or destructive effects on neurons were investigated. DPM may induce both direct neurotoxicity without exogenous stimulation and indirect neurotoxicity after Aβ activation. On the other hand, PAM attenuates Aβ-mediated neurotoxicity through Aβ phagocytosis and/or Aβ degradation. Conclusions Our results suggest that the proliferating microglia, but not the differentiated microglia, protect neurons against Aβ-mediated neurotoxicity. This discovery may be helpful on the therapeutic investigation of Alzheimer’s disease.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Young-Ji Shiao
- Institute of Biopharmaceutical Science, National Yang-Ming University, Taipei 11221, Taiwan.
| |
Collapse
|
48
|
Wang CJ, Chen CC, Tsay HJ, Chiang FY, Wu MF, Shiao YJ. Cudrania cochinchinensis attenuates amyloid β protein-mediated microglial activation and promotes glia-related clearance of amyloid β protein. J Biomed Sci 2013; 20:55. [PMID: 23915297 PMCID: PMC3750318 DOI: 10.1186/1423-0127-20-55] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Accepted: 07/31/2013] [Indexed: 01/21/2023] Open
Abstract
Background Microglial inflammation may significantly contribute to the pathology of Alzheimer’s disease. To examine the potential of Cudrania cochinchinensis to ameliorate amyloid β protein (Aβ)-induced microglia activation, BV-2 microglial cell line, and the ramified microglia in the primary glial mixed cultured were employed. Results Lipopolysaccharide (LPS), Interferon-γ (IFN-γ), fibrillary Aβ (fAβ), or oligomeric Aβ (oAβ) were used to activate microglia. LPS and IFN-γ, but not Aβs, activated BV-2 cells to produce nitric oxide through an increase in inducible nitric oxide synthase (iNOS) expression without significant effects on cell viability of microglia. fAβ, but not oAβ, enhanced the IFN-γ-stimulated nitric oxide production and iNOS expression. The ethanol/water extracts of Cudrania cochinchinensis (CC-EW) and the purified isolated components (i.e. CCA to CCF) effectively reduced the nitric oxide production and iNOS expression stimulated by IFN-γ combined with fAβ. On the other hand, oAβ effectively activated the ramified microglia in mixed glial culture by observing the morphological alteration of the microglia from ramified to amoeboid. CC-EW and CCB effectively prohibit the Aβ-mediated morphological change of microglia. Furthermore, CC-EW and CCB effectively decreased Aβ deposition and remained Aβ in the conditioned medium suggesting the effect of CC-EW and CCB on promoting Aβ clearance. Results are expressed as mean ± S.D. and were analyzed by ANOVA with post-hoc multiple comparisons with a Bonferroni test. Conclusions The components of Cudrania cochinchinensis including CC-EW and CCB are potential for novel therapeutic intervention for Alzheimer’s disease.
Collapse
Affiliation(s)
- Chung-Jen Wang
- Division of Geriatrics, Cheng Hsin General Hospital, Taipei, Taiwan
| | | | | | | | | | | |
Collapse
|
49
|
Jiang J, Dingledine R. Prostaglandin receptor EP2 in the crosshairs of anti-inflammation, anti-cancer, and neuroprotection. Trends Pharmacol Sci 2013; 34:413-23. [PMID: 23796953 DOI: 10.1016/j.tips.2013.05.003] [Citation(s) in RCA: 95] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Revised: 05/05/2013] [Accepted: 05/10/2013] [Indexed: 10/26/2022]
Abstract
Modulation of a specific prostanoid synthase or receptor provides therapeutic alternatives to nonsteroidal anti-inflammatory drugs (NSAIDs) for treating pathological conditions governed by cyclooxygenase-2 (COX-2 or PTGS2). Among the COX-2 downstream signaling pathways, the prostaglandin E2 (PGE2) receptor EP2 subtype (PTGER2) is emerging as a crucial mediator of many physiological and pathological events. Genetic ablation strategies and recent advances in chemical biology provide tools for a better understanding of EP2 signaling. In the brain, the EP2 receptor modulates some beneficial effects, including neuroprotection, in acute models of excitotoxicity, neuroplasticity, and spatial learning via cAMP-PKA signaling. Conversely, EP2 activation accentuates chronic inflammation mainly through the cAMP-Epac pathway, likely contributing to delayed neurotoxicity. EP2 receptor activation also engages β-arrestin in a G-protein-independent pathway that promotes tumor cell growth and migration. Understanding the conditions under which multiple EP2 signaling pathways are engaged might suggest novel therapeutic strategies to target this key inflammatory prostaglandin receptor.
Collapse
Affiliation(s)
- Jianxiong Jiang
- Department of Pharmacology, Emory University School of Medicine, Atlanta, GA 30322, USA.
| | | |
Collapse
|
50
|
Silencing of the P2X(7) receptor enhances amyloid-β phagocytosis by microglia. Biochem Biophys Res Commun 2013; 434:363-9. [PMID: 23562658 DOI: 10.1016/j.bbrc.2013.03.079] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2013] [Accepted: 03/26/2013] [Indexed: 11/21/2022]
Abstract
P2X7 receptor (P2X7R) is an ATP-gated cation channel that promotes microglia activation and plays a critical role in the pathogenesis of Alzheimer's disease. Inhibiting P2X7R indirectly reduces the rate of amyloid-β (Aβ)-induced neurodegeneration by suppressing secretion of inflammatory factors from activated microglia. We used RNA interference to silence P2X7R in microglial cells in vitro and found it markedly increased microglial phagocytosis of Aβ1-42. Increased phagocytic activity was dependent on decreasing the rate of interleukin-1β release from microglia and required inhibition of the COX-2 pathway. Modulation of microglial phagocytosis and secretion via silencing P2X7R may be a promising therapeutic option for the treatment of Alzheimer's disease.
Collapse
|