1
|
Zhao DP, Xue A, Yuan K, Zuo K, Li Y, Bian ZH, Xu HD, Cheng SM, Yang M, Zhao HM, Ma X, Lei X, Zhang N. Anemarrhena asphodeloides Bunge and Phellodendri Chinensis Cortex inhibits the PTGS2/EP2/cAMP/Epac1 signaling pathway to reduce microglial M1 polarization, thereby blocking chronic stress-induced depression-like behavior. JOURNAL OF ETHNOPHARMACOLOGY 2025; 347:119792. [PMID: 40216047 DOI: 10.1016/j.jep.2025.119792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Revised: 04/01/2025] [Accepted: 04/08/2025] [Indexed: 04/16/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Anemarrhena asphodeloides Bunge and Phellodendri Chinensis Cortex (AP) is a synergistic drug combination used to treat depression. However, the molecular mechanism underlying the therapeuticeffects of AP requires further elucidation. AIM OF THE STUDY To investigate the potential of AP in the treatment of depression and its mechanism of action. METHODS AND METHODS The depression model was established by chronic unpredictable mild stress (CUMS) and treated with different doses of AP. UPLC-Q-TOF-MS/MS and network pharmacology methods were used to analyze the composition and potential targets of AP. Molecular docking, affinity ultrafiltration (AUF) and cellular thermal shift assays (CETSA) techniques were used to analyze the interaction between AP components and key targets. The therapeutic effects of AP active ingredients were explored in LPS-induced cell models. RESULTS AP ameliorated CUMS-induced despair behaviors, regulate M1/M2 phenotypic balance of microglia and reduced neuroinflammation. Then, A total of 10 brain components were identified in the mPFC of CUMS rats. Based on these chemical compositions, PTGS2 was found to be at the core of the regulatory network through network analysis. All components of AP had good interaction with PTGS2, among which Neomangiferin and PTGS2 had the strongest binding capacity. AP can decrease PTGS2 activity, decrease PGE2 production and EP2 expression downstream, and reduce inflammatory response. In LPS-induced BV2 cells, Neomangiferin regulated the imbalance of M1/M2 microglia, reduced the release of pro-inflammatory factors, and decreased the expression of PTGS2, PEG2, EP2, cAMP, and Epac1. CONCLUSION AP improves the potential therapeutic benefits of depression by targeting the PTGS2 signaling pathway, thereby promoting the wider use of AP in depression.
Collapse
Affiliation(s)
- De-Ping Zhao
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, 150040, PR China
| | - Ao Xue
- Wuxi Affiliated Hospital of Nanjing University of Chinese Medicine, Wuxi, Jiangsu, 214071, PR China
| | - Ke Yuan
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, 150040, PR China
| | - Kun Zuo
- College of Pharmacy, Jiamusi University, Jiamusi, Heilongjiang, 154007, PR China
| | - Yang Li
- Jinzhou Women and Children's Hospital (Maternal and Child Health Care Hospital), Jinzhou, Liaoning, 121000, PR China
| | - Zhen-Hua Bian
- Wuxi Affiliated Hospital of Nanjing University of Chinese Medicine, Wuxi, Jiangsu, 214071, PR China
| | - Hong-Dan Xu
- Wuxi Affiliated Hospital of Nanjing University of Chinese Medicine, Wuxi, Jiangsu, 214071, PR China; College of Pharmacy, Wuxi Higher Health Vocational Technology School, Wuxi, Jiangsu, 214028, PR China
| | - Si-Min Cheng
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, 150040, PR China
| | - Meng Yang
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, 150040, PR China
| | - Hong-Mei Zhao
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, 150040, PR China
| | - Xin Ma
- College of Basic Medicine, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, 150040, PR China
| | - Xia Lei
- Wuxi Affiliated Hospital of Nanjing University of Chinese Medicine, Wuxi, Jiangsu, 214071, PR China; Jiangsu CM Clinical Innovation Center of Degenerative Bone & Joint Disease, Wuxi Affiliated Hospital of Nanjing University of Chinese Medicine, Wuxi, Jiangsu, 214071, PR China.
| | - Ning Zhang
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, 150040, PR China; Jiangsu CM Clinical Innovation Center of Degenerative Bone & Joint Disease, Wuxi Affiliated Hospital of Nanjing University of Chinese Medicine, Wuxi, Jiangsu, 214071, PR China.
| |
Collapse
|
2
|
Du X, Gao F, Chen S, Botchway BOA, Amin N, Hu Z, Fang M. Combinational Pretreatment of Colony-Stimulating Factor 1 Receptor Inhibitor and Triptolide Upregulates BDNF-Akt and Autophagic Pathways to Improve Cerebral Ischemia. Mediators Inflamm 2020; 2020:8796103. [PMID: 33192177 PMCID: PMC7648715 DOI: 10.1155/2020/8796103] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 08/27/2020] [Accepted: 09/22/2020] [Indexed: 01/15/2023] Open
Abstract
Ki20227, a selective inhibitor of colony-stimulating factor 1 receptor (CSF1R), has been suggested to regulate microglia inflammatory function and neuronal synaptic plasticity. Triptolide (TP) pretreatment has neuroprotective effects through its anti-inflammatory and antiapoptotic features in ischemic stroke mice. However, the underlying mechanism and pathway are presently unclear. We thus investigated the association between neuroprotective effects of combined TP and Ki20227 and BDNF-Akt and autophagy pathways. Ki20227 was administrated for 7 days, and TP was administered once 24 hours prior to building the ischemic stroke model in C57BL/6 mice. Behavioral tests, Golgi staining, immunofluorescence, and western blot analyses were employed to examine neuroprotective effects of TP and Ki20227. TP and Ki20227 pretreatments improved the neurobehavioral function in stroke mice. Synaptic protein expressions and density of dendritic spine density were upregulated in Ki20227 and TP pretreated stroke mice. Further, optimized integration of TP and Ki20227 pretreatments upregulated the NeuN expression and downregulated Iba1 expression after stroke. In addition, both TP and Ki20227 pretreatments significantly upregulated BDNF, p-Akt/Akt, and Erk1/2 protein expressions and autophagy related proteins (LC3II/I, Atg5, and p62), indicating the activation of BDNF and autophagic pathways. Optimized integration of TP and Ki20227 can improve cerebral ischemia by inhibiting CSF1R signal and trigger autophagy and BDNF-Akt signaling pathways to increase dendritic spine density and synaptic protein expressions, which in turn enhances neurobehavioral function.
Collapse
Affiliation(s)
- Xiaoxue Du
- Institute of Neuroscience, School of Medicine, Zhejiang University, Hangzhou, China
- Translation Medicine Center, Affiliated Hangzhou First People's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Feng Gao
- Department of Neurology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Shijia Chen
- Institute of Neuroscience, School of Medicine, Zhejiang University, Hangzhou, China
| | | | - Nashwa Amin
- Institute of Neuroscience, School of Medicine, Zhejiang University, Hangzhou, China
- Department of Zoology, Faculty of Science, Aswan University, Egypt
| | - Zhiying Hu
- Department of Obstetrics and Gynecology, Hangzhou Red Cross Hospital, Zhejiang Integrated Traditional and Western Medicine Hospital, Hangzhou, China
| | - Marong Fang
- Institute of Neuroscience, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
3
|
Porta C, Consonni FM, Morlacchi S, Sangaletti S, Bleve A, Totaro MG, Larghi P, Rimoldi M, Tripodo C, Strauss L, Banfi S, Storto M, Pressiani T, Rimassa L, Tartari S, Ippolito A, Doni A, Soldà G, Duga S, Piccolo V, Ostuni R, Natoli G, Bronte V, Balzac F, Turco E, Hirsch E, Colombo MP, Sica A. Tumor-Derived Prostaglandin E2 Promotes p50 NF-κB-Dependent Differentiation of Monocytic MDSCs. Cancer Res 2020; 80:2874-2888. [PMID: 32265223 DOI: 10.1158/0008-5472.can-19-2843] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 02/28/2020] [Accepted: 04/02/2020] [Indexed: 11/16/2022]
Abstract
Myeloid-derived suppressor cells (MDSC) include immature monocytic (M-MDSC) and granulocytic (PMN-MDSC) cells that share the ability to suppress adaptive immunity and to hinder the effectiveness of anticancer treatments. Of note, in response to IFNγ, M-MDSCs release the tumor-promoting and immunosuppressive molecule nitric oxide (NO), whereas macrophages largely express antitumor properties. Investigating these opposing activities, we found that tumor-derived prostaglandin E2 (PGE2) induces nuclear accumulation of p50 NF-κB in M-MDSCs, diverting their response to IFNγ toward NO-mediated immunosuppression and reducing TNFα expression. At the genome level, p50 NF-κB promoted binding of STAT1 to regulatory regions of selected IFNγ-dependent genes, including inducible nitric oxide synthase (Nos2). In agreement, ablation of p50 as well as pharmacologic inhibition of either the PGE2 receptor EP2 or NO production reprogrammed M-MDSCs toward a NOS2low/TNFαhigh phenotype, restoring the in vivo antitumor activity of IFNγ. Our results indicate that inhibition of the PGE2/p50/NO axis prevents MDSC-suppressive functions and restores the efficacy of anticancer immunotherapy. SIGNIFICANCE: Tumor-derived PGE2-mediated induction of nuclear p50 NF-κB epigenetically reprograms the response of monocytic cells to IFNγ toward an immunosuppressive phenotype, thus retrieving the anticancer properties of IFNγ. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/80/13/2874/F1.large.jpg.
Collapse
Affiliation(s)
- Chiara Porta
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale "Amedeo Avogadro", Novara, Italy.,Center for Translational Research on Autoimmune & Allergic Diseases (CAAD) Cso Trieste 15/A, Novara, Italy
| | | | - Sara Morlacchi
- Humanitas Clinical and Research Center - IRCCS, Rozzano, Milan, Italy
| | | | - Augusto Bleve
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale "Amedeo Avogadro", Novara, Italy
| | | | - Paola Larghi
- Humanitas Clinical and Research Center - IRCCS, Rozzano, Milan, Italy
| | - Monica Rimoldi
- Humanitas Clinical and Research Center - IRCCS, Rozzano, Milan, Italy
| | - Claudio Tripodo
- Human Pathology Section, Department of Health Sciences, University of Palermo, Palermo, Italy
| | - Laura Strauss
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale "Amedeo Avogadro", Novara, Italy
| | - Stefania Banfi
- Humanitas Clinical and Research Center - IRCCS, Rozzano, Milan, Italy
| | - Mariangela Storto
- Humanitas Clinical and Research Center - IRCCS, Rozzano, Milan, Italy
| | - Tiziana Pressiani
- Humanitas Clinical and Research Center - IRCCS, Rozzano, Milan, Italy
| | - Lorenza Rimassa
- Humanitas Clinical and Research Center - IRCCS, Rozzano, Milan, Italy
| | - Silvia Tartari
- Humanitas Clinical and Research Center - IRCCS, Rozzano, Milan, Italy
| | - Alessandro Ippolito
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale "Amedeo Avogadro", Novara, Italy
| | - Andrea Doni
- Humanitas Clinical and Research Center - IRCCS, Rozzano, Milan, Italy
| | - Giulia Soldà
- Humanitas Clinical and Research Center - IRCCS, Rozzano, Milan, Italy.,Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | - Stefano Duga
- Humanitas Clinical and Research Center - IRCCS, Rozzano, Milan, Italy.,Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | - Viviana Piccolo
- Humanitas Clinical and Research Center - IRCCS, Rozzano, Milan, Italy
| | - Renato Ostuni
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, Milan, Italy.,Vita-Salute San Raffaele University, Milan, Italy
| | - Gioacchino Natoli
- Humanitas Clinical and Research Center - IRCCS, Rozzano, Milan, Italy.,Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | - Vincenzo Bronte
- Department of Medicine, Verona University Hospital, Verona, Italy
| | - Fiorella Balzac
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Emilia Turco
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Emilio Hirsch
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Mario P Colombo
- Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Antonio Sica
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale "Amedeo Avogadro", Novara, Italy. .,Humanitas Clinical and Research Center - IRCCS, Rozzano, Milan, Italy
| |
Collapse
|
4
|
Wang L, Liu Y, Yan S, Du T, Fu X, Gong X, Zhou X, Zhang T, Wang X. Disease Progression-Dependent Expression of CD200R1 and CX3CR1 in Mouse Models of Parkinson's Disease. Aging Dis 2020; 11:254-268. [PMID: 32257540 PMCID: PMC7069458 DOI: 10.14336/ad.2019.0615] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 06/15/2019] [Indexed: 12/16/2022] Open
Abstract
Microglial activation is an important contributor to the pathogenesis of Parkinson’s disease (PD). Microglia are tightly and efficiently regulated by immune checkpoints, including CD200-CD200R1 and CX3CL1-CX3CR1. Understanding the involvement of these checkpoints in disease progression provides important insights into how microglial activation contributes to PD pathology. However, so far, studies have produced seemingly conflicting results. In this study, we demonstrate that CD200R1 expression is down-regulated at both early and late stage of PD model, and CX3CR1 expression is down-regulated in early stage and recovered in late stage. In primary cultured microglia, CD200R1 and CX3CR1 expressions are both directly regulated by LPS or α-synuclein, and CD200R1 expression is more sensitively regulated than CX3CR1. In addition, CD200 knockout causes an increase in proinflammatory cytokine production and microglial activation in the midbrain. Remarkably, DA neurons in the substantial nigra are degenerated in CD200-/- mice. Finally, activation of the CD200R with CD200Fc alleviates the neuroinflammation in microglia. Together, these results suggest that immune checkpoints play distinct functional roles in different stage of PD pathology, and the CD200-CD200R1 axis plays a significant role in nigrostriatal neuron viability and function.
Collapse
Affiliation(s)
- Le Wang
- 1Department of Neurobiology, Center of Parkinson Disease Beijing Institute for Brain Disorders, Beijing Key Laboratory on Parkinson Disease, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China
| | - Yang Liu
- 1Department of Neurobiology, Center of Parkinson Disease Beijing Institute for Brain Disorders, Beijing Key Laboratory on Parkinson Disease, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China
| | - Shuxin Yan
- 1Department of Neurobiology, Center of Parkinson Disease Beijing Institute for Brain Disorders, Beijing Key Laboratory on Parkinson Disease, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China
| | - Tianshu Du
- 1Department of Neurobiology, Center of Parkinson Disease Beijing Institute for Brain Disorders, Beijing Key Laboratory on Parkinson Disease, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China
| | - Xia Fu
- 1Department of Neurobiology, Center of Parkinson Disease Beijing Institute for Brain Disorders, Beijing Key Laboratory on Parkinson Disease, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China
| | - Xiaoli Gong
- 2Department of Physiology and Pathophysiology, Capital Medical University, Beijing, China
| | - Xinyu Zhou
- 1Department of Neurobiology, Center of Parkinson Disease Beijing Institute for Brain Disorders, Beijing Key Laboratory on Parkinson Disease, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China
| | - Ting Zhang
- 1Department of Neurobiology, Center of Parkinson Disease Beijing Institute for Brain Disorders, Beijing Key Laboratory on Parkinson Disease, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China
| | - Xiaomin Wang
- 1Department of Neurobiology, Center of Parkinson Disease Beijing Institute for Brain Disorders, Beijing Key Laboratory on Parkinson Disease, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China.,2Department of Physiology and Pathophysiology, Capital Medical University, Beijing, China
| |
Collapse
|
5
|
Wang L, Gong X, Liu Y, Du T, Zhang Z, Zhang T, Wang X. CD200 maintains the region-specific phenotype of microglia in the midbrain and its role in Parkinson's disease. Glia 2020; 68:1874-1890. [PMID: 32112601 DOI: 10.1002/glia.23811] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Revised: 01/23/2020] [Accepted: 02/18/2020] [Indexed: 12/21/2022]
Abstract
Microglia are a specialized population of tissue macrophages in the mammalian brain. Microglial phenotype is tightly regulated by local environmental factors, although little is known about these factors and their region-preferred roles in regulating local neuroinflammatory responses. We hypothesized that microglia in different brain regions respond differently to neuroinflammatory stimulation and that CD200, an anti-inflammatory protein mainly originated from neurons, acts as a local cue inhibiting microglia activation in the midbrain. We utilized a CD200-deficient mouse line to analyze the phenotypic role of CD200 in the regulation of normal neuron-microglia homeostasis in the midbrain and in the dopaminergic degeneration in an α-synuclein overexpression model of PD. We found that systemic administration of an endotoxin lipopolysaccharide induced a region-preferred change in CD200 expression in the midbrain. Similarly, CD200-/- mice showed a regional preference in an enhancement of microglia activation and baseline inflammatory levels in the midbrain and dopamine neuron loss in the substantia nigra (SN). In a mouse model of Parkinson's disease (PD) induced by rAAV-hSYN injection into the SN, CD200-/- mice showed more dopamine neuron loss in the SN than wild type mice. Activation of CD200 receptors with a CD200 fusion protein alleviated the neuroinflammation and neuronal death in the SN of PD mice. These findings demonstrate that CD200 is essential for the midbrain homeostasis and acts as a critical local regulator in controlling microglial properties related to the PD pathogenesis.
Collapse
Affiliation(s)
- Le Wang
- Department of Neurobiology, Center of Parkinson Disease Beijing Institute for Brain Disorders, Beijing Key Laboratory on Parkinson Disease, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China
| | - Xiaoli Gong
- Department of Physiology and Pathophysiology, Capital Medical University, Beijing, China
| | - Yang Liu
- Department of Neurobiology, Center of Parkinson Disease Beijing Institute for Brain Disorders, Beijing Key Laboratory on Parkinson Disease, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China
| | - Tianshu Du
- Department of Neurobiology, Center of Parkinson Disease Beijing Institute for Brain Disorders, Beijing Key Laboratory on Parkinson Disease, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China
| | - Zhen Zhang
- Department of Neurobiology, Center of Parkinson Disease Beijing Institute for Brain Disorders, Beijing Key Laboratory on Parkinson Disease, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China
| | - Ting Zhang
- Department of Neurobiology, Center of Parkinson Disease Beijing Institute for Brain Disorders, Beijing Key Laboratory on Parkinson Disease, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China
| | - Xiaomin Wang
- Department of Neurobiology, Center of Parkinson Disease Beijing Institute for Brain Disorders, Beijing Key Laboratory on Parkinson Disease, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China
| |
Collapse
|
6
|
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: 94] [Impact Index Per Article: 15.7] [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
|
7
|
Triptolide Suppressed the Microglia Activation to Improve Spinal Cord Injury Through miR-96/IKKβ/NF-κB Pathway. Spine (Phila Pa 1976) 2019; 44:E707-E714. [PMID: 31150368 DOI: 10.1097/brs.0000000000002989] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
STUDY DESIGN The effect of triptolide on spinal cord injury (SCI) and inflammatory response was observed by establishing SCI rat model. And in vitro experiments were conducted to determine the underlying mechanism of triptolide-mediated in murine microglial cell line BV2. OBJECTIVE To determine the underlying mechanism of triptolide in suppressing the microglia activation to improve SCI. SUMMARY OF BACKGROUND DATA Triptolide, as a major active ingredient of Chinese herb Tripterygium wilfordii, can promote spinal cord repair through inhibiting microglia activation, but the underlying mechanism is not clear. METHODS Locomotion recovery was accessed by Basso, Beattie, and Bresnahan score, the number of footfalls, stride length, and angle of rotation analysis. Expressions of microRNA 96 (miR-96), microglia activation marker Iba-1, and IκB kinase (IKKβ)/nuclear factor (NF)-κB-related proteins were detected by qRT-PCR or western blot. Inflammatory cytokines tumor necrosis factor-α and interleukin -1β were measured by enzyme-linked immuno sorbent assay. The regulation of miR-96 on IKKβ was confirmed by dual luciferase reporter assay. RESULTS Triptolide promoted locomotion recovery of SCI rats, upregulated the expression of miR-96, decreased microglia activation marker Iba-1 and IKKβ/NF-κB-related proteins, and inhibited inflammatory cytokines tumor necrosis factor-α and interleukin-1β levels in spinal cord tissues and lipopolysaccharide -induced microglia. Triptolide suppressed the microglia activation and inflammatory cytokines secretion in BV2 cells through up-regulating miR-96. We confirmed the interaction between miR-96 and IKKβ, and IKKβ expression was negatively regulated by miR-96. Finally, we determined that triptolide suppressed the microglia activation and inflammatory cytokines secretion through miR-96/IKKβ pathway. CONCLUSION Triptolide suppressed microglia activation after SCI through miR-96/IKKβ/NF-κB pathway. LEVEL OF EVIDENCE N/A.
Collapse
|
8
|
Li J, Hao J. Treatment of Neurodegenerative Diseases with Bioactive Components of Tripterygium wilfordii. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2019; 47:769-785. [PMID: 31091976 DOI: 10.1142/s0192415x1950040x] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Tripterygium wilfordii Hook F. (TWHF), a traditional Chinese medicine, has been widely used to treat autoimmune and inflammatory diseases including rheumatoid arthritis, systemic lupus erythematosus and dermatomyositis in China. Recently, studies have demonstrated that the bioactive components of TWHF have effective therapeutic potential for neurodegenerative diseases including Alzheimer's disease, Parkinson's disease and Multiple Sclerosis. In this paper, we summarize the research progress of triptolide and celastrol (the two major TWHF components) as well as their analogues in the treatment of neurodegenerative diseases. In addition, we review and discuss the molecular mechanisms and structure features of those two bioactive TWHF components, highlighting their therapeutic promise in neurodegenerative diseases.
Collapse
Affiliation(s)
- Jianheng Li
- * School of Pharmacy, Key laboratory of Pharmaceutical Quality Control of Hebei Province, Hebei University, Baoding, Hebei, P. R. China
| | - Jijun Hao
- † College of Veterinary Medicine, Western University of Health Sciences, Pomona, CA 91766, USA.,‡ Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766, USA
| |
Collapse
|
9
|
Distinct influence of COX-1 and COX-2 on neuroinflammatory response and associated cognitive deficits during high altitude hypoxia. Neuropharmacology 2019; 146:138-148. [DOI: 10.1016/j.neuropharm.2018.11.026] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 10/26/2018] [Accepted: 11/16/2018] [Indexed: 02/08/2023]
|
10
|
Wang L, Xu D, Li L, Xing X, Liu L, Ismail Abdelmotalab M, Xiao L, Pang T, Huang X, Wang X, Wang T, Jiang Z, Zhang L, Sun L. Possible role of hepatic macrophage recruitment and activation in triptolide-induced hepatotoxicity. Toxicol Lett 2018; 299:32-39. [DOI: 10.1016/j.toxlet.2018.08.017] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2018] [Revised: 07/16/2018] [Accepted: 08/27/2018] [Indexed: 12/22/2022]
|
11
|
Low, but not high, dose triptolide controls neuroinflammation and improves behavioral deficits in toxic model of multiple sclerosis by dampening of NF-κB activation and acceleration of intrinsic myelin repair. Toxicol Appl Pharmacol 2018; 342:86-98. [DOI: 10.1016/j.taap.2018.01.023] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2017] [Revised: 01/15/2018] [Accepted: 01/29/2018] [Indexed: 12/12/2022]
|
12
|
Chen SR, Dai Y, Zhao J, Lin L, Wang Y, Wang Y. A Mechanistic Overview of Triptolide and Celastrol, Natural Products from Tripterygium wilfordii Hook F. Front Pharmacol 2018; 9:104. [PMID: 29491837 PMCID: PMC5817256 DOI: 10.3389/fphar.2018.00104] [Citation(s) in RCA: 214] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 01/30/2018] [Indexed: 12/28/2022] Open
Abstract
Triptolide and celastrol are predominantly active natural products isolated from the medicinal plant Tripterygium wilfordii Hook F. These compounds exhibit similar pharmacological activities, including anti-cancer, anti-inflammation, anti-obesity, and anti-diabetic activities. Triptolide and celastrol also provide neuroprotection and prevent cardiovascular and metabolic diseases. However, toxicity restricts the further development of triptolide and celastrol. In this review, we comprehensively review therapeutic targets and mechanisms of action, and translational study of triptolide and celastrol. We systemically discuss the structure-activity-relationship of triptolide, celastrol, and their derivatives. Furthermore, we propose the use of structural derivatives, targeted therapy, and combination treatment as possible solutions to reduce toxicity and increase therapeutic window of these potent natural products from T. wilfordii Hook F.
Collapse
Affiliation(s)
- Shao-Ru Chen
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Yan Dai
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Jing Zhao
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Ligen Lin
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Yitao Wang
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Ying Wang
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macau, China
| |
Collapse
|
13
|
Sasmita AO, Ling APK, Voon KGL, Koh RY, Wong YP. Madecassoside activates anti‑neuroinflammatory mechanisms by inhibiting lipopolysaccharide‑induced microglial inflammation. Int J Mol Med 2018; 41:3033-3040. [PMID: 29436598 DOI: 10.3892/ijmm.2018.3479] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 01/08/2018] [Indexed: 11/06/2022] Open
Abstract
Neurodegeneration is typically preceded by neuroinflammation generated by the nervous system to protect itself from tissue damage, however, excess neuroinflammation may inadvertently cause more harm to the surrounding tissues. Attenuating neuroinflammation with non‑steroidal anti‑inflammatory drugs can inhibit neurodegeneration. However, such treatments induce chronic side effects, including stomach ulcers. Madecassoside, a triterpene derived from Centella asiatica, is considered to be an alternative treatment of inflammation. In the present study, the anti‑neuroinflammatory properties of madecassoside were assessed in BV2 microglia cells, which were pre‑treated with madecassoside at a maximum non‑toxic dose (MNTD) of 9.50 µg/ml and a ½ MNTD of 4.75 µg/ml for 3 h and stimulated with 0.1 µg/ml lipopolysaccharide (LPS). The effect of madecassoside was assessed by determining reactive oxygen species (ROS) levels in all groups. Furthermore, the expression of pro‑ and anti‑neuroinflammatory genes and proteins were analyzed using reverse transcription‑quantitative polymerase chain reaction and western blotting, respectively. The results demonstrated that ROS levels in cells treated with the MNTD of madecassoside were significantly reduced compared with cells treated with LPS alone (P<0.05). The expression of pro‑neuroinflammatory genes, including inducible nitric oxide synthase, cyclooxygenase‑2, signal transducer and activator of transcription 1 and nuclear factor‑κB, were significantly downregulated in a dose‑independent manner following treatment with madecassoside. Conversely, the anti‑neuroinflammatory component heme oxygenase 1 was significantly upregulated by 175.22% in the MNTD‑treated group, compared with cells treated with LPS alone (P<0.05). The gene expression profiles of pro‑ and anti‑inflammatory genes were also consistent with the results of western blotting. The results of the present study suggest that madecassoside may be a potent anti‑neuroinflammatory agent. The antioxidative properties of madecassoside, which serve a major role in anti‑neuroinflammation, indicate that this compound may be a functional natural anti‑neuroinflammatory agent, therefore, further in vivo or molecular studies are required.
Collapse
Affiliation(s)
- Andrew Octavian Sasmita
- Division of Applied Biomedical Sciences and Biotechnology, School of Health Sciences, International Medical University, Kuala Lumpur 57000, Malaysia
| | - Anna Pick Kiong Ling
- Division of Applied Biomedical Sciences and Biotechnology, School of Health Sciences, International Medical University, Kuala Lumpur 57000, Malaysia
| | - Kenny Gah Leong Voon
- Division of Pathology, School of Medicine, International Medical University, Kuala Lumpur 57000, Malaysia
| | - Rhun Yian Koh
- Division of Applied Biomedical Sciences and Biotechnology, School of Health Sciences, International Medical University, Kuala Lumpur 57000, Malaysia
| | - Ying Pei Wong
- Division of Applied Biomedical Sciences and Biotechnology, School of Health Sciences, International Medical University, Kuala Lumpur 57000, Malaysia
| |
Collapse
|
14
|
Wang J, Qiao Y, Yang RS, Zhang CK, Wu HH, Lin JJ, Zhang T, Chen T, Li YQ, Dong YL, Li JL. The synergistic effect of treatment with triptolide and MK-801 in the rat neuropathic pain model. Mol Pain 2017; 13:1744806917746564. [PMID: 29166839 PMCID: PMC5734437 DOI: 10.1177/1744806917746564] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Triptolide (T10), an active component of Tripterygium wilfordii Hook F, is reported to have potent anti-inflammatory and analgesic effects. Additionally, MK-801, a noncompetitive N-methyl-D-aspartate receptor antagonist, can reduce glutamate toxicity and has a significant analgesic effect on chronic pain. In this study, we tested the possible synergistic analgesic ability by intrathecal administration of T10 and MK-801 for the treatment of neuropathic pain. Single T10 (3, 10, or 30 µg/kg), MK-801 (10, 30, or 90 µg/kg), or a combination of them were intrathecally administrated in rats with spinal nerve ligation. We found that single administration of T10 caused a slow-acting but long-term analgesic effect, while single administration of MK-801 caused a fast-acting but short-term effect. Administration of their combination showed obviously synergic analgesia and the 1:3 ratio of T10 to MK-801 reached the peak effect. Furthermore, application of T10 and/or MK-801 significantly inhibited the activation of microglia and astrocyte and phosphorylation of STAT3 and NR2B in the spinal dorsal horn induced by chronic neuropathic pain. Our data suggest that the combination of T10 and MK-801 may be a potentially novel strategy for treatment of neuropathic pain.
Collapse
Affiliation(s)
- Jian Wang
- 1 Department of Anatomy and K. K. Leung Brain Research Centre, Fourth Military Medical University, Xi'an, China.,2 Student Brigade, Fourth Military Medical University, Xi'an, China
| | - Yu Qiao
- 1 Department of Anatomy and K. K. Leung Brain Research Centre, Fourth Military Medical University, Xi'an, China.,2 Student Brigade, Fourth Military Medical University, Xi'an, China
| | - Ri-Sheng Yang
- 1 Department of Anatomy and K. K. Leung Brain Research Centre, Fourth Military Medical University, Xi'an, China.,2 Student Brigade, Fourth Military Medical University, Xi'an, China
| | - Chun-Kui Zhang
- 1 Department of Anatomy and K. K. Leung Brain Research Centre, Fourth Military Medical University, Xi'an, China
| | - Huang-Hui Wu
- 1 Department of Anatomy and K. K. Leung Brain Research Centre, Fourth Military Medical University, Xi'an, China.,3 Department of Anesthesiology, Fuzhou General Hospital of Nanjing Military Region, Fuzhou, China
| | - Jia-Ji Lin
- 1 Department of Anatomy and K. K. Leung Brain Research Centre, Fourth Military Medical University, Xi'an, China.,4 Department of Neurology, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Ting Zhang
- 1 Department of Anatomy and K. K. Leung Brain Research Centre, Fourth Military Medical University, Xi'an, China
| | - Tao Chen
- 1 Department of Anatomy and K. K. Leung Brain Research Centre, Fourth Military Medical University, Xi'an, China
| | - Yun-Qing Li
- 1 Department of Anatomy and K. K. Leung Brain Research Centre, Fourth Military Medical University, Xi'an, China.,5 Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Yu-Lin Dong
- 1 Department of Anatomy and K. K. Leung Brain Research Centre, Fourth Military Medical University, Xi'an, China
| | - Jin-Lian Li
- 1 Department of Anatomy and K. K. Leung Brain Research Centre, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
15
|
Liu SP, Wang GD, Du XJ, Wan G, Wu JT, Miao LB, Liang QD. Triptolide inhibits the function of TNF-α in osteoblast differentiation by inhibiting the NF-κB signaling pathway. Exp Ther Med 2017; 14:2235-2240. [PMID: 28962148 DOI: 10.3892/etm.2017.4749] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 01/20/2017] [Indexed: 12/15/2022] Open
Abstract
Chronic inflammation often delays fracture healing or leads to bone nonunion. Effectively suppressing pathological inflammation is crucial for fracture healing or bone remodeling. Triptolide, which is a diterpenoid epoxide, is the major active component of the Thunder God Vine, Tripterygium wilfordii. The aim of the present study was to investigate the role of triptolide in osteoblast differentiation and explore the molecular mechanisms of triptolide in fracture healing. Alkaline phosphatase (ALP) activity was used to evaluate osteoblast differentiation. ALP activity was measured via histochemical staining and western blotting was used to determine the expression of factors associated with inflammation. C2C12 cells were initially treated with 200 ng/ml bone morphogenetic protein (BMP)-2 alone for 3 days, which caused a significant increase in ALP activity (P<0.01). However, treatment with tumor necrosis factor (TNF)-α significantly decreased the ALP activity (P<0.05). Notably, treatment with the chronic inflammatory cytokine TNF-α significantly decreased the effect of BMP-2 in C2C12 cells compared with BMP-2 treatment alone (P<0.01). C2C12 cells were treated with increasing concentrations of BMP-2 or TNF-α for 3 days. The results demonstrated that TNF-α treatment significantly inhibited BMP-2-induced osteoblast differentiation in a dose-dependent manner (P<0.01). The role of triptolide in BMP-2-induced osteoblast differentiation was also examined. Cells were treated with BMP-2, BMP-2 + TNF-α alone, or BMP2 + TNF-α with increasing concentrations of triptolide (4, 8 or 16 ng/ml). After 3 days, the results of ALP activity revealed that triptolide significantly reversed the TNF-α-associated inhibition of osteoblast differentiation (P<0.01). Western blotting analysis demonstrated that triptolide markedly inhibited the phosphorylation of nuclear factor-κB, therefore suppressing the effects of TNF-α. In summary, triptolide is able to reverse the TNF-α-associated suppression of osteoblast differentiation, suggesting that triptolide treatment may have a positive effect on bone remodeling and fracture repairing.
Collapse
Affiliation(s)
- Shen-Peng Liu
- Department of Orthopedics, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, P.R. China
| | - Guo-Dong Wang
- Department of Orthopedics, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, P.R. China
| | - Xue-Jun Du
- Department of Orthopedics, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, P.R. China
| | - Guang Wan
- Department of Orthopedics, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, P.R. China
| | - Jun-Tao Wu
- Department of Orthopedics, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Lian-Bao Miao
- Department of Orthopedics, Huaxian People's Hospital of Henan Province, Anyang, Henan 456400, P.R. China
| | - Qiu-Dong Liang
- Department of Orthopedics, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, P.R. China
| |
Collapse
|
16
|
An EP2 Agonist Facilitates NMDA-Induced Outward Currents and Inhibits Dendritic Beading through Activation of BK Channels in Mouse Cortical Neurons. Mediators Inflamm 2016; 2016:5079597. [PMID: 27298516 PMCID: PMC4889853 DOI: 10.1155/2016/5079597] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Revised: 04/19/2016] [Accepted: 05/04/2016] [Indexed: 11/17/2022] Open
Abstract
Prostaglandin E2 (PGE2), a major metabolite of arachidonic acid produced by cyclooxygenase pathways, exerts its bioactive responses by activating four E-prostanoid receptor subtypes, EP1, EP2, EP3, and EP4. PGE2 enables modulating N-methyl-D-aspartate (NMDA) receptor-mediated responses. However, the effect of E-prostanoid receptor agonists on large-conductance Ca2+-activated K+ (BK) channels, which are functionally coupled with NMDA receptors, remains unclear. Here, we showed that EP2 receptor-mediated signaling pathways increased NMDA-induced outward currents (INMDA-OUT), which are associated with the BK channel activation. Patch-clamp recordings from the acutely dissociated mouse cortical neurons revealed that an EP2 receptor agonist activated INMDA-OUT, whereas an EP3 receptor agonist reduced it. Agonists of EP1 or EP4 receptors showed no significant effects on INMDA-OUT. A direct perfusion of 3,5′-cyclic adenosine monophosphate (cAMP) through the patch pipette facilitated INMDA-OUT, which was abolished by the presence of protein kinase A (PKA) inhibitor. Furthermore, facilitation of INMDA-OUT caused by an EP2 receptor agonist was significantly suppressed by PKA inhibitor. Finally, the activation of BK channels through EP2 receptors facilitated the recovery phase of NMDA-induced dendritic beading in the primary cultured cortical neurons. These results suggest that a direct activation of BK channels by EP2 receptor-mediated signaling pathways plays neuroprotective roles in cortical neurons.
Collapse
|
17
|
Davidson JM, Wong CT, Rai-Bhogal R, Li H, Crawford DA. Prostaglandin E2 elevates calcium in differentiated neuroectodermal stem cells. Mol Cell Neurosci 2016; 74:71-7. [PMID: 27074429 DOI: 10.1016/j.mcn.2016.03.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Revised: 03/21/2016] [Accepted: 03/31/2016] [Indexed: 01/02/2023] Open
Abstract
Lipid mediator prostaglandin E2 (PGE2) is an endogenous signaling molecule that plays an important role during early development of the nervous system. Abnormalities in the PGE2 signaling pathway have been associated with neurodevelopmental disorders such as autism spectrum disorders. In this study we use ratiometric fura-2AM calcium imaging to show that higher levels of PGE2 elevate intracellular calcium levels in the cell soma and growth cones of differentiated neuroectodermal (NE-4C) stem cells. PGE2 also increased the amplitude of calcium fluctuation in the neuronal growth cones and affected the neurite extension length. In summary, our results show that PGE2 may adversely impact intracellular calcium dynamics in differentiated neuronal cells and possibly affect early development of the nervous system.
Collapse
Affiliation(s)
- Jennilee M Davidson
- Neuroscience Graduate Diploma Program, York University, Toronto, ON M3J 1P3, Canada; Department of Biology, York University, Toronto, ON M3J 1P3, Canada
| | - Christine T Wong
- School of Kinesiology and Health Science, York University, Toronto, ON M3J 1P3, Canada; Neuroscience Graduate Diploma Program, York University, Toronto, ON M3J 1P3, Canada
| | - Ravneet Rai-Bhogal
- Neuroscience Graduate Diploma Program, York University, Toronto, ON M3J 1P3, Canada; Department of Biology, York University, Toronto, ON M3J 1P3, Canada
| | - Hongyan Li
- School of Kinesiology and Health Science, York University, Toronto, ON M3J 1P3, Canada
| | - Dorota A Crawford
- School of Kinesiology and Health Science, York University, Toronto, ON M3J 1P3, Canada; Neuroscience Graduate Diploma Program, York University, Toronto, ON M3J 1P3, Canada; Department of Biology, York University, Toronto, ON M3J 1P3, Canada.
| |
Collapse
|
18
|
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: 1.6] [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
|