1
|
Berdiaki A, Thrapsanioti LN, Giatagana EM, K Karamanos N, C Savani R, N Tzanakakis G, Nikitovic D. RHAMM/hyaluronan inhibit β-catenin degradation, enhance downstream signaling, and facilitate fibrosarcoma cell growth. Mol Biol Rep 2023; 50:8937-8947. [PMID: 37710072 DOI: 10.1007/s11033-023-08763-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 08/16/2023] [Indexed: 09/16/2023]
Abstract
Increased hyaluronan deposition (HA) in various cancer tissues, including sarcomas, correlates with disease progression. The receptor for hyaluronic acid-mediated motility (RHAMM) expression is elevated in most human cancers. β-catenin is a critical downstream mediator of the Wnt signaling pathways, facilitating carcinogenic events characterized by deregulated cell proliferation. We previously showed that low molecular weight (LMW) HA/RHAMM/β-catenin signaling axis increases HT1080 fibrosarcoma cell growth. Here, focusing on mechanistic aspects and utilizing immunofluorescence and immunoprecipitation, we demonstrate that LMW HA treatment enhanced RHAMM intracellular localization (p ≤ 0.001) and RHAMM/β-catenin colocalization in HT1080 fibrosarcoma cells (p ≤ 0.05). Downregulating endogenous HA attenuated the association of RHAMM/β-catenin in HT1080 fibrosarcoma cells (p ≤ 0.0.01). Notably, Axin-2, the key β-catenin degradation complex component, and RHAMM were demonstrated to form a complex primarily to cell membranes, enhanced by LMW HA (p ≤ 0.01). In contrast, LMW HA attenuated the association of β-catenin and Axin-2 (p ≤ 0.05). The utilization of FH535, a Wnt signaling inhibitor, showed that LMW HA partially rescued the Wnt-dependent growth of HT1080 cells and restored the expression of Wnt/β-catenin mediators, cyclin-D1 and c-myc (p ≤ 0.05). B6FS fibrosarcoma cells with different HA metabolism do not respond to the LMW HA growth stimulus (p = NS). The present study identifies a novel LMW HA/RHAMM mechanism in a fibrosarcoma model. LMW HA regulates intracellular RHAMM expression, which acts as a scaffold protein binding β-catenin and Axin-2 at different cellular compartments to increase β-catenin expression, transcriptional activity, and fibrosarcoma growth.
Collapse
Affiliation(s)
- Aikaterini Berdiaki
- Laboratory of Histology-Embryology, School of Medicine, University of Crete, Heraklion, 71003, Greece
| | - Lydia-Nefeli Thrapsanioti
- Laboratory of Histology-Embryology, School of Medicine, University of Crete, Heraklion, 71003, Greece
| | - Eirini-Maria Giatagana
- Laboratory of Histology-Embryology, School of Medicine, University of Crete, Heraklion, 71003, Greece
| | | | - Rashmin C Savani
- Department of Pediatrics, University of Florida College of Medicine, 1600 SW Archer Road, P.O. Box 100296, Gainesville, FL, USA
| | - George N Tzanakakis
- Laboratory of Histology-Embryology, School of Medicine, University of Crete, Heraklion, 71003, Greece
| | - Dragana Nikitovic
- Laboratory of Histology-Embryology, School of Medicine, University of Crete, Heraklion, 71003, Greece.
| |
Collapse
|
2
|
Li C, Wu Y, Huang MY, Song XJ. Characterization of Inflammatory Signals in BV-2 Microglia in Response to Wnt3a. Biomedicines 2023; 11:biomedicines11041121. [PMID: 37189739 DOI: 10.3390/biomedicines11041121] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 03/31/2023] [Accepted: 04/05/2023] [Indexed: 05/17/2023] Open
Abstract
Activation of microglia is one of the pathological bases of neuroinflammation, which involves various diseases of the central nervous system. Inhibiting the inflammatory activation of microglia is a therapeutic approach to neuroinflammation. In this study, we report that activation of the Wnt/β-catenin signaling pathway in a model of neuroinflammation in Lipopolysaccharide (LPS)/IFN-γ-stimulated BV-2 cells can result in inhibition of production of nitric oxide (NO), interleukin-6 (IL-6), and tumor necrosis factor-α (TNF-α). Activation of the Wnt/β-catenin signaling pathway also results in inhibition of the phosphorylation of nuclear factor-κB (NF-κB) and extracellular signal-regulated kinase (ERK) in the LPS/IFN-γ-stimulated BV-2 cells. These findings indicate that activation of the Wnt/β-catenin signaling pathway can inhibit neuroinflammation through downregulating the pro-inflammatory cytokines including iNOS, TNF-α, and IL-6, and suppress NF-κB/ERK-related signaling pathways. In conclusion, this study indicates that the Wnt/β-catenin signaling activation may play an important role in neuroprotection in certain neuroinflammatory diseases.
Collapse
Affiliation(s)
- Cheng Li
- Department of Medical Neuroscience, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China
- SUSTech Center for Pain Medicine, Southern University of Science and Technology, Shenzhen 518055, China
| | - Ying Wu
- Department of Medical Neuroscience, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China
- SUSTech Center for Pain Medicine, Southern University of Science and Technology, Shenzhen 518055, China
| | - Ming-Yue Huang
- Department of Medical Neuroscience, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China
| | - Xue-Jun Song
- Department of Medical Neuroscience, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China
- SUSTech Center for Pain Medicine, Southern University of Science and Technology, Shenzhen 518055, China
| |
Collapse
|
3
|
Alomar HA, Nadeem A, Ansari MA, Attia SM, Bakheet SA, Al-Mazroua HA, Alhazzani K, Assiri MA, Alqinyah M, Almudimeegh S, Ahmad SF. Mitogen-activated protein kinase inhibitor PD98059 improves neuroimmune dysfunction in experimental autoimmune encephalomyelitis in SJL/J mice through the inhibition of nuclear factor-kappa B signaling in B cells. Brain Res Bull 2023; 194:45-53. [PMID: 36646144 DOI: 10.1016/j.brainresbull.2023.01.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Revised: 12/20/2022] [Accepted: 01/12/2023] [Indexed: 01/15/2023]
Abstract
Multiple sclerosis (MS) is a severe autoimmune disease leading to demyelination, followed by consequent axonal degeneration, causing sensory, motor, cognitive, and visual symptoms. Experimental autoimmune encephalomyelitis (EAE) is the most well-studied animal model of MS. Most current MS treatments are not completely effective, and severe side effects remain a great challenge. In this study, we report the therapeutic efficacy of PD98059, a potent mitogen-activated protein kinase inhibitor, on proteolipid protein (PLP)139-151-induced EAE in SJL/J mice. Following the induction of EAE, mice were intraperitoneally treated with PD98059 (5 mg/kg for 14 days) daily from day 14 to day 28. This study investigated the effects of PD98059 on C-C motif chemokine receptor 6 (CCR6), CD14, NF-κB p65, IκBα, GM-CSF, iNOS, IL-6, TNF-α in CD45R+ B lymphocytes using flow cytometry. Furthermore, we analyzed the effect of PD98059 on CCR6, CD14, NF-κB p65, GM-CSF, iNOS, IL-6, and TNF-α mRNA and protein expression levels using qRT-PCR analysis in brain tissues. Mechanistic investigations revealed that PD98059-treated in mice with EAE had reduced CD45R+CCR6+, CD45R+CD14+, CD45R+NF-κB p65+, CD45R+GM-CSF+, CD45R+iNOS+, CD45R+IL-6+, and CD45R+TNF-α+ cells and increased CD45R+IκBα+ cells compared with vehicle-treated control mice in the spleen. Moreover, downregulation of CCR6, CD14, NF-κB p65, GM-CSF, iNOS, IL-6, and TNF-α mRNA expression level was observed in PD98059-treated mice with EAE compared with vehicle-treated control mice in the brain tissue. The results of this study demonstrate that PD98059 modulates inflammatory mediators through multiple cellular mechanisms. The results of this study suggest that PD98059 may be pursued as a therapeutic agent for the treatment of MS.
Collapse
Affiliation(s)
- Hatun A Alomar
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Ahmed Nadeem
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mushtaq A Ansari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Sabry M Attia
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Saleh A Bakheet
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Haneen A Al-Mazroua
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Khalid Alhazzani
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohammed A Assiri
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohammed Alqinyah
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Sultan Almudimeegh
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Sheikh F Ahmad
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia.
| |
Collapse
|
4
|
Kwon J, Arsenis C, Suessmilch M, McColl A, Cavanagh J, Morris BJ. Differential Effects of Toll-Like Receptor Activation and Differential Mediation by MAP Kinases of Immune Responses in Microglial Cells. Cell Mol Neurobiol 2021; 42:2655-2671. [PMID: 34297254 PMCID: PMC9560989 DOI: 10.1007/s10571-021-01127-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 07/10/2021] [Indexed: 10/26/2022]
Abstract
Microglial activation is believed to play a role in many psychiatric and neurodegenerative diseases. Based largely on evidence from other cell types, it is widely thought that MAP kinase (ERK, JNK and p38) signalling pathways contribute strongly to microglial activation following immune stimuli acting on toll-like receptor (TLR) 3 or TLR4. We report here that exposure of SimA9 mouse microglial cell line to immune mimetics stimulating TLR4 (lipopolysaccharide-LPS) or TLR7/8 (resiquimod/R848), results in marked MAP kinase activation, followed by induction of nitric oxide synthase, and various cytokines/chemokines. However, in contrast to TLR4 or TLR7/8 stimulation, very few effects of TLR3 stimulation by poly-inosine/cytidine (polyI:C) were detected. Induction of chemokines/cytokines at the mRNA level by LPS and resiquimod were, in general, only marginally affected by MAP kinase inhibition, and expression of TNF, Ccl2 and Ccl5 mRNAs, along with nitrite production, were enhanced by p38 inhibition in a stimulus-specific manner. Selective JNK inhibition enhanced Ccl2 and Ccl5 release. Many distinct responses to stimulation of TLR4 and TLR7 were observed, with JNK mediating TNF protein induction by the latter but not the former, and suppressing Ccl5 release by the former but not the latter. These data reveal complex modulation by MAP kinases of microglial responses to immune challenge, including a dampening of some responses. They demonstrate that abnormal levels of JNK or p38 signalling in microglial cells will perturb their profile of cytokine and chemokine release, potentially contributing to abnormal inflammatory patterns in CNS disease states.
Collapse
Affiliation(s)
- Jaedeok Kwon
- Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of Glasgow, West Medical Building, Glasgow, G12 8QQ, UK.,Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Christos Arsenis
- Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of Glasgow, West Medical Building, Glasgow, G12 8QQ, UK
| | - Maria Suessmilch
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Alison McColl
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Jonathan Cavanagh
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Brian J Morris
- Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of Glasgow, West Medical Building, Glasgow, G12 8QQ, UK.
| |
Collapse
|
5
|
Katarzyna Greda A, Nowicka D. Hyaluronidase inhibition accelerates functional recovery from stroke in the mouse brain. J Neurochem 2021; 157:781-801. [PMID: 33345310 DOI: 10.1111/jnc.15279] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 11/30/2020] [Accepted: 12/17/2020] [Indexed: 12/15/2022]
Abstract
Perineuronal nets (PNNs) are presumed to limit plasticity in adult animals. Ischaemic stroke results in the massive breakdown of PNNs resulting in rejuvenating states of neuronal plasticity, but the mechanisms of this phenomenon are largely unknown. As hyaluronic acid (HA) is the structural backbone of PNNs, we hypothesized that these changes are a consequence of the altered expression of HA metabolism enzymes. Additionally, we investigated whether early hyaluronidase inhibition interferes with post-stroke PNN reduction and behavioural recovery. We investigated the mRNA/protein expression of these enzymes in the perilesional, remote and contralateral cortical regions in mice at different time points after photothrombosis, using quantitative real-time polymerase chain reaction and immunofluorescence. A skilled reaching test was employed to test hyaluronidase inhibitor L-ascorbic acid 6-hexadecanoate influence on post-stroke recovery. We found the simultaneous up-regulation of mRNA of HA synthesizing and degrading enzymes in the perilesional area early after stroke, suggesting an acceleration of HA turnover in ischaemic animals. Immunostaining revealed differential cellular localization of enzymes, with hyaluronidase 1 in astrocytes and hyaluronan synthase 2 in astrocytes and neurons, and post-stroke up-regulation of both of them in astrocytes. β-glucuronidase was observed in neurons but post-stroke up-regulation occurred in microglia. Inhibition of hyaluronidase activity early after stroke resulted in improved performance in skilled reaching test, without affecting the numbers of PNNs. These results suggest that after stroke, a substantial reorganization of polysaccharide content occurs, and interfering with this process at early time has a beneficial effect on recovery.
Collapse
Affiliation(s)
- Anna Katarzyna Greda
- Nencki Institute of Experimental Biology PAS, Laboratory of Epileptogenesis, Warsaw, Poland
| | - Dorota Nowicka
- Nencki Institute of Experimental Biology PAS, Laboratory of Epileptogenesis, Warsaw, Poland
| |
Collapse
|
6
|
Yao Y, Fu KY. Serum-deprivation leads to activation-like changes in primary microglia and BV-2 cells but not astrocytes. Biomed Rep 2020; 13:51. [PMID: 32974017 DOI: 10.3892/br.2020.1358] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 07/28/2020] [Indexed: 12/22/2022] Open
Abstract
The aim of the present study was to determine the effect of serum deprivation on primary microglia, BV-2 cells and primary astrocytes. Cell morphology combined with the expression of phospho-(p-)38 and p-extracellular signal-regulated kinase (ERK) were assessed. Serum deprivation resulted in various alterations in the three cell cultures. Primary microglia and BV-2 cells exhibited alterations indicative of activation under serum treatment, as well as lipopolysaccharide (LPS) treatment. However, astrocytes did not react as fast. Regarding morphology, the processes present on the primary microglia and BV-2 cells became shorter and the cell bodies became larger, and more transparent vesicles were observed within the cell bodies, which indicated their increased phagocytic ability. At the protein level, p-p38 expression increased quickly within 1 h in the primary microglia culture in response to LPS treatment. Furthermore, the expression levels of p-p38 and p-ERK were elevated in both primary microglia and BV-2 cells under serum deprivation, as well as under LPS treatment, which was not observed in the primary astrocytes. These results suggest that serum deprivation may result in similar changes to cell morphology and the expression levels of p-p38 and p-ERK as LPS treatment in primary microglia and BV-2 cells. These observations suggest that primary microglia and BV-2 cells may become activated under serum deprivation, at least to a certain degree.
Collapse
Affiliation(s)
- Yao Yao
- Department of Oral and Maxillofacial Surgery, Beijing Stomatological Hospital and School of Stomatology, Capital Medical University, Beijing 100050, P.R. China
| | - Kai-Yuan Fu
- Center for TMD and Orofacial Pain, Peking University School and Hospital of Stomatology, Beijing 100081, P.R. China
| |
Collapse
|
7
|
Liu J, Li H, Gong T, Chen W, Mao S, Kong Y, Yu J, Sun J. Anti-neuroinflammatory Effect of Short-Chain Fatty Acid Acetate against Alzheimer's Disease via Upregulating GPR41 and Inhibiting ERK/JNK/NF-κB. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:7152-7161. [PMID: 32583667 DOI: 10.1021/acs.jafc.0c02807] [Citation(s) in RCA: 111] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Alzheimer's disease (AD) is a high-incidence neurodegenerative disease in the elderly. Acetate (Ace) is a short-chain fatty acid (SCFA) with neuroprotective activity. The purpose of this study was to investigate the effects and its possible mechanisms of SCFA Ace on AD. A male APP/PS1 transgenic mouse was given intragastric administration Ace for 4 weeks. Cognitive function and microglia activation in mice were assessed. Furthermore, Ace pretreated amyloid-β (Aβ)-induced BV2 microglia, and the levels of CD11b, COX-2, and G-protein-coupled receptor 41 (GPR41) and phosphorylation of ERK, JNK, and NF-κB p65 were determined. Our results revealed that Ace significantly attenuated the cognitive impairment and decreased the CD11b level in the APP/PS1 mice. Moreover, Ace inhibited the phosphorylation of NF-κB p65, ERK, and JNK and decreased the levels of COX-2 and interleukin 1β in the Aβ-stimulated BV2 microglia. Finally, Ace increased the GPR41 level in the Aβ-stimulated BV2 cells. The finding indicated that Ace exerted antineuroinflammatory effects via the upregulation of GPR41 and suppression of the ERK/JNK/NF-κB pathway, which might provide an alternative therapy strategy of AD.
Collapse
Affiliation(s)
- Jiaming Liu
- Department of Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, People's Republic of China
- Department of Preventive Medicine, School of Public Health and Management, Wenzhou Medical University, Wenzhou, Zhejiang 325035, People's Republic of China
| | - Haijun Li
- Department of Neurology, Taizhou Second People's Hospital, Taizhou, Zhejiang 317000, People's Republic of China
| | - Tianyu Gong
- Department of Preventive Medicine, School of Public Health and Management, Wenzhou Medical University, Wenzhou, Zhejiang 325035, People's Republic of China
| | - Wenyang Chen
- Department of Preventive Medicine, School of Public Health and Management, Wenzhou Medical University, Wenzhou, Zhejiang 325035, People's Republic of China
| | - Shiyin Mao
- Department of Preventive Medicine, School of Public Health and Management, Wenzhou Medical University, Wenzhou, Zhejiang 325035, People's Republic of China
| | - Yu Kong
- Department of Preventive Medicine, School of Public Health and Management, Wenzhou Medical University, Wenzhou, Zhejiang 325035, People's Republic of China
| | - Jiaheng Yu
- Department of Preventive Medicine, School of Public Health and Management, Wenzhou Medical University, Wenzhou, Zhejiang 325035, People's Republic of China
| | - Jing Sun
- Department of Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, People's Republic of China
| |
Collapse
|
8
|
Zhu L, Liu X, Nemeth DP, DiSabato DJ, Witcher KG, Mckim DB, Oliver B, Le X, Gorantla G, Berdysz O, Li J, Ramani AD, Chen Z, Wu D, Godbout JP, Quan N. Interleukin-1 causes CNS inflammatory cytokine expression via endothelia-microglia bi-cellular signaling. Brain Behav Immun 2019; 81:292-304. [PMID: 31228609 PMCID: PMC6754782 DOI: 10.1016/j.bbi.2019.06.026] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 05/26/2019] [Accepted: 06/18/2019] [Indexed: 12/30/2022] Open
Abstract
As a major producer of the inflammatory cytokine interleukin-1 (IL-1), peripheral macrophages can augment IL-1 expression via type 1 IL-1 receptor (IL-1R1) mediated autocrine self-amplification. In the CNS, microglial cells are the major producers of inflammatory cytokines, but express negligible levels of IL-1R1. In the present study, we showed CNS IL-1 induced microglial proinflammatory cytokine expression was mediated by endothelial, not microglial, IL-1R1. This paracrine mechanism was further dissected in vitro. IL-1 was unable to stimulate inflammatory cytokine expression directly from the microglial cell line BV-2, but it stimulated the brain endothelial cell line bEnd.3 to produce a factor(s) in the culture supernatant, which was capable of inducing inflammatory cytokine expression in BV-2. We termed this factor IL-1-induced microglial activation factors (IMAF). BV-2 cytokine expression was inducible by extracellular ATP, but IL-1 did not stimulate the release of ATP from bEnd.3 cells. Filtration of IMAF by size-exclusion membranes showed IMAF activity resided in molecules larger than 50 kd and incubation of IMAF at 95 °C for 5 min did not alter its activity. Microglial inhibitor minocycline was unable to block IMAF activity, even though it blocked LPS induced cytokine expression in BV-2 cells. Adding NF-κB inhibitor to the bEnd.3 cells abolished IL-1 induced cytokine expression in this bi-cellular system, but adding NF-κB inhibitor after IMAF is already produced failed to abrogate IMAF induced cytokine expression in BV-2 cells. RNA sequencing of IL-1 stimulated endothelial cells revealed increased expression of genes involved in the production and processing of hyaluronic acid (HA), suggesting HA as a candidate of IMAF. Inhibition of hyaluronidase by ascorbyl palmitate (AP) abolished IMAF-induced cytokine expression in BV-2 cells. AP administration in vivo also inhibited ICV IL-1-induced IL-1 expression in the hippocampus and hypothalamus. In vitro, either TLR2 or TLR4 inhibitors blocked IMAF induced BV-2 cytokine expression. In vivo, however, IL-1 induced cytokine expression persisted in either TLR2 or TLR4 knockouts. These results demonstrate IL-1 induced inflammatory cytokine expression in the CNS requires a bi-cellular system and HA could be a candidate for IMAF.
Collapse
Affiliation(s)
- Ling Zhu
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, 610041, P.R.China
| | - Xiaoyu Liu
- Department of Biomedical Science, Charles E. Schmidt College of Medicine and Brain Institute, Florida Atlantic University, Jupiter, FL 33458, USA.
| | - Daniel P. Nemeth
- Department of Biomedical Science, Charles E. Schmidt College of Medicine and Brain Institute, Florida Atlantic University, Jupiter, FL 33458, USA, Institute for Behavioral Medicine Research, College of Medicine, The Ohio State University, Columbus, OH 43210, USA, Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH 43210, USA
| | - Damon J. DiSabato
- Institute for Behavioral Medicine Research, College of Medicine, The Ohio State University, Columbus, OH 43210, USA, Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH 43210, USA, Department of Neuroscience, The Ohio State University, Columbus, OH 43210, USA
| | - Kristina G. Witcher
- Institute for Behavioral Medicine Research, College of Medicine, The Ohio State University, Columbus, OH 43210, USA, Department of Neuroscience, The Ohio State University, Columbus, OH 43210, USA
| | - Daniel B. Mckim
- Department of Animal Science, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Braedan Oliver
- Institute for Behavioral Medicine Research, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Xi Le
- Department of Biomedical Science, Charles E. Schmidt College of Medicine and Brain Institute, Florida Atlantic University, Jupiter, FL 33458, USA, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei 430072, P.R.China, Wuhan Hamilton Biotechnology Co., Ltd., Wuhan, Hubei 430075, P.R.China
| | - Gowthami Gorantla
- Institute for Behavioral Medicine Research, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Olimpia Berdysz
- Institute for Behavioral Medicine Research, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Jiaoni Li
- Institute for Behavioral Medicine Research, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Aishwarya D. Ramani
- Institute for Behavioral Medicine Research, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Zhibiao Chen
- Department of Neurosurgery, Renmin Hospital, Wuhan University, Wuhan, Hubei 430060, P.R.China
| | - Dongcheng Wu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei 430072, P.R.China, Wuhan Hamilton Biotechnology Co., Ltd., Wuhan, Hubei 430075, P.R.China
| | - Jonathan P. Godbout
- Institute for Behavioral Medicine Research, College of Medicine, The Ohio State University, Columbus, OH 43210, USA, Department of Neuroscience, The Ohio State University, Columbus, OH 43210, USA
| | - Ning Quan
- Department of Biomedical Science, Charles E. Schmidt College of Medicine and Brain Institute, Florida Atlantic University, Jupiter, FL 33458, USA.
| |
Collapse
|
9
|
Lin SS, Yuan LJ, Niu CC, Tu YK, Yang CY, Ueng SWN. Hyperbaric oxygen inhibits the HMGB1/RAGE signaling pathway by upregulating Mir-107 expression in human osteoarthritic chondrocytes. Osteoarthritis Cartilage 2019; 27:1372-1381. [PMID: 31146014 DOI: 10.1016/j.joca.2019.05.011] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 05/15/2019] [Accepted: 05/17/2019] [Indexed: 02/02/2023]
Abstract
OBJECTIVE MicroRNA (miRNA)107 expression is downregulated but high mobility group box 1 (HMGB-1), Toll-like receptors (TLRs), and receptor for advanced glycation end products (RAGE) are upregulated in osteoarthritic (OA) cartilage. We investigated mir-107/HMGB-1 signaling in OA after hyperbaric oxygen (HBO) treatment. DESIGN MiR-107 mimic was transfected and the HMGB-1 was analyzed in OA chondrocytes. MiRNA targets were identified using bioinformatics and a luciferase reporter assay. After HBO treatment, the mRNA or protein levels of HMGB-1, RAGE, TLR2, TLR4, and inducible nitric oxide (NO) synthase (iNOS) and phosphorylation of mitogen-activated protein kinase (MAPK) were evaluated. The secreted HMGB-1 and matrix metalloproteases (MMPs) levels were quantified. Finally, we detected the HMGB-1 and iNOS expression in rabbit cartilage defects. RESULTS Overexpression of miR-107 suppressed HMGB-1 expression in OA chondrocytes. The 3'UTR of HMGB-1 mRNA contained a 'seed-matched-sequence' for miR-107. MiR-107 was induced by HBO and a marked suppression of HMGB-1 was observed simultaneously in OA chondrocytes. Knockdown of miR-107 upregulated HMGB-1 expression in hyperoxic cells. HBO downregulated the mRNA and protein expression of HMGB-1, RAGE, TLR2, TLR4, and iNOS, and the secretion of HMGB-1. HBO decreased the nuclear translocation of nuclear factor (NF)-κB, downregulated the phosphorylation of MAPK, and significantly decreased the secretion of MMPs. Morphological and immunohistochemical observation demonstrated that HBO markedly enhanced cartilage repair and the area stained positive for HMGB-1 and iNOS tended to be lower in the HBO group. CONCLUSIONS HBO inhibits HMGB-1/RAGE signaling related pathways by upregulating miR-107 expression in human OA chondrocytes.
Collapse
Affiliation(s)
- S-S Lin
- Department of Orthopaedic Surgery, Chang Gung Memorial Hospital, Linkou, Taiwan; Department of Nursing, Chang Gung University of Science and Technology, Taoyuan, Taiwan.
| | - L-J Yuan
- Department of Orthopaedic Surgery, E-Da Hospital, I-Shou University.
| | - C-C Niu
- Department of Orthopaedic Surgery, Chang Gung Memorial Hospital, Linkou, Taiwan.
| | - Y-K Tu
- Department of Orthopaedic Surgery, E-Da Hospital, I-Shou University.
| | - C-Y Yang
- Department of Orthopaedic Surgery, Chang Gung Memorial Hospital, Linkou, Taiwan.
| | - S W N Ueng
- Department of Orthopaedic Surgery, Chang Gung Memorial Hospital, Linkou, Taiwan; College of Medicine, Chang Gung University, Taoyuan, Taiwan; Bone and Joint Research Center, Chang Gung Memorial Hospital, Linkou, Taiwan.
| |
Collapse
|
10
|
Niu CC, Lin SS, Yuan LJ, Lu ML, Ueng SWN, Yang CY, Tsai TT, Lai PL. Upregulation of miR-107 expression following hyperbaric oxygen treatment suppresses HMGB1/RAGE signaling in degenerated human nucleus pulposus cells. Arthritis Res Ther 2019; 21:42. [PMID: 30704538 PMCID: PMC6357369 DOI: 10.1186/s13075-019-1830-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 01/18/2019] [Indexed: 02/07/2023] Open
Abstract
Background The expression of both high-mobility group box 1 (HMGB1) and receptor for advanced glycation end-products (RAGE) is upregulated in degenerated discs. HMGB1 is known to function as a coupling factor between hypoxia and inflammation in arthritis, and this inflammatory response is modulated by microRNAs (miRNAs), with miR-107 expression downregulated during hypoxia. In this study, we investigated the regulation of the miR-107/HMGB1/RAGE pathway in degenerated nucleus pulposus cells (NPCs) after hyperbaric oxygen (HBO) treatment. Methods NPCs were separated from human degenerated intervertebral disc tissues. The control cells were maintained in 5% CO2/95% air, and the hyperoxic cells were exposed to 100% O2 at 2.5 atmospheres absolute. MiRNA expression profiling was performed via microarray and confirmed by real-time PCR, and miRNA target genes were identified using bioinformatics and luciferase reporter assays. The cellular protein and mRNA levels of HMGB1, RAGE, and inducible nitric oxide synthase (iNOS) were assessed, and the phosphorylation of MAPK (p38MAPK, ERK, and JNK) was evaluated. Additionally, cytosolic and nuclear fractions of the IκBα and NF-κB p65 proteins were analyzed, and secreted HMGB1 and metalloprotease (MMP) levels in the conditioned media were quantified. Results Using microarray analyses, 96 miRNAs were identified as upregulated and 66 downregulated following HBO treatment. Based on these results, miR-107 was selected for further investigation. Bioinformatics analyses indicated that the 3′ untranslated region of the HMGB1 mRNA contained the “seed-matched-sequence” for hsa-miR-107, which was validated via dual-luciferase reporter assays. MiR-107 was markedly induced by HBO, and simultaneous suppression of HMGB1 was observed in NPCs. Knockdown of miR-107 resulted in upregulation of HMGB1 expression in HBO-treated cells, and HBO treatment downregulated the mRNA and protein levels of HMGB1, RAGE, and iNOS and the secretion of HMGB1. In addition, HBO treatment upregulated the protein levels of cytosolic IκBα and decreased the nuclear translocation of NF-κB in NPCs. Moreover, HBO treatment downregulated the phosphorylation of p38MAPK, ERK, and JNK and significantly decreased the secretion of MMP-3, MMP-9, and MMP-13. Conclusions HBO inhibits pathways related to HMGB1/RAGE signaling via upregulation of miR-107 expression in degenerated human NPCs.
Collapse
Affiliation(s)
- Chi-Chien Niu
- Department of Orthopaedic Surgery, Chang Gung Memorial Hospital, No 5, Fu-Hsing Street 333, Taoyuan, Taoyuan, Taiwan. .,College of Medicine, Chang Gung University, Taoyuan, Taiwan.
| | - Song-Shu Lin
- Department of Orthopaedic Surgery, Chang Gung Memorial Hospital, No 5, Fu-Hsing Street 333, Taoyuan, Taoyuan, Taiwan.,Department of Nursing, Chang Gung University of Science and Technology, Taoyuan, Taiwan
| | - Li-Jen Yuan
- Department of Orthopaedic Surgery, E-Da Hospital/I-Shou University, Kaohsiung, Taiwan
| | - Meng-Ling Lu
- Department of Orthopaedic Surgery, Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Steve W N Ueng
- Department of Orthopaedic Surgery, Chang Gung Memorial Hospital, No 5, Fu-Hsing Street 333, Taoyuan, Taoyuan, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chuen-Yung Yang
- Department of Orthopaedic Surgery, Chang Gung Memorial Hospital, No 5, Fu-Hsing Street 333, Taoyuan, Taoyuan, Taiwan
| | - Tsung-Ting Tsai
- Department of Orthopaedic Surgery, Chang Gung Memorial Hospital, No 5, Fu-Hsing Street 333, Taoyuan, Taoyuan, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Bone and Joint Research Center, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Po-Liang Lai
- Department of Orthopaedic Surgery, Chang Gung Memorial Hospital, No 5, Fu-Hsing Street 333, Taoyuan, Taoyuan, Taiwan
| |
Collapse
|
11
|
Majors AK, Chakravarti R, Ruple LM, Leahy R, Stuehr DJ, Lauer M, Erzurum SC, Janocha A, Aronica MA. Nitric oxide alters hyaluronan deposition by airway smooth muscle cells. PLoS One 2018; 13:e0200074. [PMID: 29966020 PMCID: PMC6028120 DOI: 10.1371/journal.pone.0200074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 06/19/2018] [Indexed: 12/04/2022] Open
Abstract
Asthma is a chronic inflammatory disease that is known to cause changes in the extracellular matrix, including changes in hyaluronan (HA) deposition. However, little is known about the factors that modulate its deposition or the potential consequences. Asthmatics with high levels of exhaled nitric oxide (NO) are characterized by greater airway reactivity and greater evidence of airway inflammation. Based on these data and our previous work we hypothesized that excessive NO promotes the pathologic production of HA by airway smooth muscle cells (SMCs). Exposure of cultured SMCs to various NO donors results in the accumulation of HA in the form of unique, cable-like structures. HA accumulates rapidly after exposure to NO and can be seen as early as one hour after NO treatment. The cable-like HA in NO-treated SMC cultures supports the binding of leukocytes. In addition, NO produced by murine macrophages (RAW cells) and airway epithelial cells also induces SMCs to produce HA cables when grown in co-culture. The modulation of HA by NO appears to be independent of soluble guanylate cyclase. Taken together, NO-induced production of leukocyte-binding HA by SMCs provides a new potential mechanism for the non-resolving airway inflammation in asthma and suggests a key role of non-immune cells in driving the chronic inflammation of the submucosa. Modulation of NO, HA and the consequent immune cell interactions may serve as potential therapeutic targets in asthma.
Collapse
Affiliation(s)
- Alana K. Majors
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Ritu Chakravarti
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Lisa M. Ruple
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Rachel Leahy
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Dennis J. Stuehr
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Mark Lauer
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Serpil C. Erzurum
- Respiratory Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Allison Janocha
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Mark A. Aronica
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
- Respiratory Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| |
Collapse
|
12
|
Lim HS, Kim YJ, Kim BY, Park G, Jeong SJ. The Anti-neuroinflammatory Activity of Tectorigenin Pretreatment via Downregulated NF-κB and ERK/JNK Pathways in BV-2 Microglial and Microglia Inactivation in Mice With Lipopolysaccharide. Front Pharmacol 2018; 9:462. [PMID: 29867470 PMCID: PMC5954245 DOI: 10.3389/fphar.2018.00462] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2017] [Accepted: 04/19/2018] [Indexed: 12/14/2022] Open
Abstract
The activation of microglia is decisively involved with the neurodegeneration observed in many neuroinflammatory pathologies, such as multiple sclerosis, Parkinson’s disease, and Alzheimer’s disease. Tectorigenin (TEC) is an isoflavone isolated from various medicinal plants, such as Pueraria thunbergiana Benth, Belamcanda chinensis, and Iris unguicularis. In the present study, the neuroinflammatory effects of TEC were evaluated in both lipopolysaccharide (LPS)-treated BV-2 microglial and mouse models. TEC remarkably inhibited reactive oxygen species (ROS) generation. TEC also inhibits the production and expression of nitric oxide (NO), prostaglandin E2 (PGE2), tumor necrosis factor-α (TNF-α), and interleukin-6 (IL-6) in LPS-stimulated BV-2 cells. In addition, TEC suppressed the LPS-induced activation of nuclear factor-κB (NF-κB), phosphorylation of extracellular signal-regulated kinase (ERK), and c-Jun N-terminal kinase (JNK) to regulate the inflammatory mediators, such as inducible NO synthase (iNOS), cyclooxygenase-2 (COX-2), TNF-α, and IL-6. These results indicate that TEC may inhibit neuronal inflammation through the downregulation of inflammatory mediators, including iNOS, COX-2, TNF-α, and IL-6 by suppressing NF-κB/ERK/JNK-related signaling pathways. Furthermore, cotreatment with TEC and ERK inhibitor SCH772984 or JNK inhibitor SP600125 suppressed the overproduction of LPS-induced NO production in BV-2 cells. Consistent with the results of in vitro experiments, an LPS-induced brain inflammation mouse model, administration of TEC effectively decrease the levels of malondialdehyde, iNOS in hippocampus, and prevented increases in the levels of TNF-α and IL-6 in the serum. TEC showed marked attenuation of microglial activation. Finally, TEC inhibited protein expression of toll-like receptor 4 and myeloid differentiation factor 88 in LPS-activated BV-2 microglia and mouse models. Taken altogether, the cumulative findings suggested that TEC holds the potential to develop as a neuroprotective drug for the intervention of neuroinflammatory disorders.
Collapse
Affiliation(s)
- Hye-Sun Lim
- Herbal Medicine Research Division, Korea Institute of Oriental Medicine, Daejeon, South Korea
| | - Yu Jin Kim
- Herbal Medicine Research Division, Korea Institute of Oriental Medicine, Daejeon, South Korea.,College of Pharmacy, Chungnam National University, Daejeon, South Korea
| | - Bu-Yeo Kim
- Herbal Medicine Research Division, Korea Institute of Oriental Medicine, Daejeon, South Korea
| | | | - Soo-Jin Jeong
- Herbal Medicine Research Division, Korea Institute of Oriental Medicine, Daejeon, South Korea.,Korean Medicine Life Science, University of Science & Technology, Daejeon, South Korea
| |
Collapse
|
13
|
Nagy N, Kuipers HF, Marshall PL, Wang E, Kaber G, Bollyky PL. Hyaluronan in immune dysregulation and autoimmune diseases. Matrix Biol 2018; 78-79:292-313. [PMID: 29625181 DOI: 10.1016/j.matbio.2018.03.022] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 03/10/2018] [Accepted: 03/30/2018] [Indexed: 02/06/2023]
Abstract
The tissue microenvironment contributes to local immunity and to the pathogenesis of autoimmune diseases - a diverse set of conditions characterized by sterile inflammation, immunity against self-antigens, and destruction of tissues. However, the specific factors within the tissue microenvironment that contribute to local immune dysregulation in autoimmunity are poorly understood. One particular tissue component implicated in multiple autoimmune diseases is hyaluronan (HA), an extracellular matrix (ECM) polymer. HA is abundant in settings of chronic inflammation and contributes to lymphocyte activation, polarization, and migration. Here, we first describe what is known about the size, amount, and distribution of HA at sites of autoimmunity and in associated lymphoid structures in type 1 diabetes, multiple sclerosis, and rheumatoid arthritis. Next, we examine the recent literature on HA and its impact on adaptive immunity, particularly in regards to the biology of lymphocytes and Foxp3+ regulatory T-cells (Treg), a T-cell subset that maintains immune tolerance in healthy individuals. We propose that HA accumulation at sites of chronic inflammation creates a permissive environment for autoimmunity, characterized by CD44-mediated inhibition of Treg expansion. Finally, we address potential tools and strategies for targeting HA and its receptor CD44 in chronic inflammation and autoimmunity.
Collapse
Affiliation(s)
- Nadine Nagy
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA.
| | - Hedwich F Kuipers
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Payton L Marshall
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Esther Wang
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Gernot Kaber
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Paul L Bollyky
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
14
|
Martini AC, Forner S, Koepp J, Rae GA. Inhibition of spinal c-Jun-NH2-terminal kinase (JNK) improves locomotor activity of spinal cord injured rats. Neurosci Lett 2016; 621:54-61. [DOI: 10.1016/j.neulet.2016.04.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Revised: 03/23/2016] [Accepted: 04/08/2016] [Indexed: 01/04/2023]
|
15
|
Electromagnetic pulse activated brain microglia via the p38 MAPK pathway. Neurotoxicology 2016; 52:144-9. [DOI: 10.1016/j.neuro.2015.12.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Revised: 12/07/2015] [Accepted: 12/08/2015] [Indexed: 11/17/2022]
|
16
|
Jiang T, Cadenas E. Astrocytic metabolic and inflammatory changes as a function of age. Aging Cell 2014; 13:1059-67. [PMID: 25233945 PMCID: PMC4244278 DOI: 10.1111/acel.12268] [Citation(s) in RCA: 131] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/09/2014] [Indexed: 12/17/2022] Open
Abstract
This study examines age-dependent metabolic-inflammatory axis in primary astrocytes isolated from brain cortices of 7-, 13-, and 18-month-old Sprague–Dawley male rats. Astrocytes showed an age-dependent increase in mitochondrial oxidative metabolism respiring on glucose and/or pyruvate substrates; this increase in mitochondrial oxidative metabolism was accompanied by increases in COX3/18SrDNA values, thus suggesting an enhanced mitochondrial biogenesis. Enhanced mitochondrial respiration in astrocytes limits the substrate supply from astrocytes to neurons; this may be viewed as an adaptive mechanism to altered cellular inflammatory–redox environment with age. These metabolic changes were associated with an age-dependent increase in hydrogen peroxide generation (largely ascribed to an enhanced expression of NOX2) and NFκB signaling in the cytosol as well as its translocation to the nucleus. Astrocytes also displayed augmented responses with age to inflammatory cytokines, IL-1β, and TNFα. Activation of NFκB signaling resulted in increased expression of nitric oxide synthase 2 (inducible nitric oxide synthase), leading to elevated nitric oxide production. IL-1β and TNFα treatment stimulated mitochondrial oxidative metabolism and mitochondrial biogenesis in astrocytes. It may be surmised that increased mitochondrial aerobic metabolism and inflammatory responses are interconnected and support the functionality switch of astrocytes, from neurotrophic to neurotoxic with age.
Collapse
Affiliation(s)
- Tianyi Jiang
- Pharmacology and Pharmaceutical Sciences School of Pharmacy University of Southern California Los Angeles CA 90089‐9121 USA
| | - Enrique Cadenas
- Pharmacology and Pharmaceutical Sciences School of Pharmacy University of Southern California Los Angeles CA 90089‐9121 USA
| |
Collapse
|
17
|
Kim HK. Role of ERK/MAPK signalling pathway in anti-inflammatory effects of Ecklonia cava in activated human mast cell line-1 cells. ASIAN PAC J TROP MED 2014. [DOI: 10.1016/s1995-7645(14)60120-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
|
18
|
Niu CC, Lin SS, Yuan LJ, Chen LH, Wang IC, Tsai TT, Lai PL, Chen WJ. Hyperbaric oxygen treatment suppresses MAPK signaling and mitochondrial apoptotic pathway in degenerated human intervertebral disc cells. J Orthop Res 2013; 31:204-9. [PMID: 22886767 DOI: 10.1002/jor.22209] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2012] [Accepted: 07/16/2012] [Indexed: 02/04/2023]
Abstract
Nucleus pulposus cells (NPCs) from degenerating discs produce catabolic and inflammatory factors, including interleukin (IL)-1 and nitric oxide (NO). Enhanced production of NO has been implicated in the apoptosis of degenerating disc cells. This study evaluates the effects of hyperbaric oxygen (HBO) on degenerated human NPCs. All hyperoxic cells were exposed to 100% O(2) at 2.5 atmospheres absolute (ATA). Phosphorylation of extracellular signal-regulated kinase 1/2 (ERK1/2), c-Jun N-terminal kinase (JNK), and p38 mitogen-activated protein kinase (MAPK) in NPCs was detected using the phosphor-kinase array kit. RNA was isolated for real-time polymerase chain reaction (PCR) analysis of aggrecan and type II collagen gene expression. The levels of IL- 1β and NO were quantified by enzyme-linked immunosorbent assay (ELISA). To identify the HBO-induced anti-apoptotic pathways, expression of Bcl-2 and Bax proteins as well as activation of cysteine-containing aspartate-specific proteases (caspases) 3, 8, and 9 was evaluated using Western blotting after HBO treatment. Our data showed that HBO treatment decreased the expression of IL-1β, suppressed phosphorylation of ERK1/2, JNK, and p38 MAPK, decreased synthesis of NO, and increased the gene expression of aggrecan and type II collagen in NPCs as compared with the atmospheric treatment. HBO up-regulated the ratio of Bcl-2 to Bax expression and reduced the activity of caspases 9 and 3 but not of caspase 8, indicating a selective effect over the mitochondrial apoptosis pathway in degenerated NPCs. These results support our hypothesis that HBO treatment suppresses MAPK signaling and mitochondrial apoptotic pathway in degenerated human intervertebral disc cells.
Collapse
Affiliation(s)
- Chi-Chien Niu
- Department of Orthopaedic Surgery, Hyperbaric Oxygen Therapy Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Piao JH, Wang Y, Duncan ID. CD44 is required for the migration of transplanted oligodendrocyte progenitor cells to focal inflammatory demyelinating lesions in the spinal cord. Glia 2012; 61:361-7. [DOI: 10.1002/glia.22438] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2012] [Accepted: 09/25/2012] [Indexed: 11/06/2022]
|
20
|
Kim SJ, Kim JS, Choi IY, Kim DH, Kim MC, An HJ, Na HJ, Kim NH, Moon PD, Myung NY, Lee JY, Jeong HJ, Um JY, Shin TY, Kim HM, Hong SH. Anti-Inflammatory Activity ofSchizonepeta tenuifoliathrough the Inhibition of MAPK Phosphorylation in Mouse Peritoneal Macrophages. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2012; 36:1145-58. [DOI: 10.1142/s0192415x0800648x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Schizonepeta tenuifolia (ST) is a well-known herb to treat the cold and its associated headache. However, the anti-inflammatory mechanism of ST in mouse peritoneal macrophages is not clear. In this study, we demonstrated that ST inhibited lipopolysaccaride (LPS)-induced tumor necrosis factor (TNF)-α and interleukin (IL)-6 production. The maximal inhibition rate of TNF-α and IL-6 production by ST (2 mg/ml) was 48.01 ± 2.8% and 56.45 ± 2.8%, respectively. During the inflammatory process, cyclooxygenase (COX)-2 and inducible nitric oxide synthase (iNOS) were increased in mouse peritoneal macrophages. However, treated with ST decreased the protein level of COX-2 and iNOS, as well as the production of PGE2and NO in LPS-stimulated mouse peritoneal macrophages. In addition, ST inhibited the phosphorylation of MAPK. Taken together, the results of this study suggest an important molecular mechanism by which ST reduces inflammation, which may explain its beneficial effect in the regulation of inflammatory reactions.
Collapse
Affiliation(s)
- Su-Jin Kim
- College of Pharmacy, Wonkwang University, Iksan, Jeonbuk, 570-749, Republic of Korea
- College of Oriental Medicine, Institute of Oriental Medicine, Kyung Hee University, 1 Hoegi-Dong, Dongdaemun-Gu, Seoul, 130-701, Republic of Korea
| | - Jung-Sun Kim
- College of Pharmacy, Wonkwang University, Iksan, Jeonbuk, 570-749, Republic of Korea
| | - In-Young Choi
- College of Pharmacy, Wonkwang University, Iksan, Jeonbuk, 570-749, Republic of Korea
| | - Dong-Hyun Kim
- College of Pharmacy, Wonkwang University, Iksan, Jeonbuk, 570-749, Republic of Korea
| | - Min-Cheol Kim
- College of Pharmacy, Wonkwang University, Iksan, Jeonbuk, 570-749, Republic of Korea
| | - Hyo-Jin An
- College of Oriental Medicine, Institute of Oriental Medicine, Kyung Hee University, 1 Hoegi-Dong, Dongdaemun-Gu, Seoul, 130-701, Republic of Korea
| | - Ho-Jeong Na
- College of Oriental Medicine, Institute of Oriental Medicine, Kyung Hee University, 1 Hoegi-Dong, Dongdaemun-Gu, Seoul, 130-701, Republic of Korea
| | - Na-Hyung Kim
- College of Oriental Medicine, Institute of Oriental Medicine, Kyung Hee University, 1 Hoegi-Dong, Dongdaemun-Gu, Seoul, 130-701, Republic of Korea
| | - Phil-Dong Moon
- Oriental Medical Science Center, College of Oriental Medicine, Kyung Hee University, 1 Hoegi-Dong, Dongdaemun-Gu, Seoul, 130-701, Republic of Korea
| | - Noh-Yil Myung
- Acupuncture and Meridian Science Research Center, Kyung Hee University, 1 Hoegi-Dong, Dongdaemun-Gu, Seoul, 130-701, Republic of Korea
| | - Ju-Young Lee
- Department of Herb Science, ShinSung College, Chungnam 343-861, Republic of Korea
| | - Hyun-Ja Jeong
- Biochip Research Center, Hoseo University, Asan, Chungnam, 336-795, Republic of Korea
| | - Jae-Young Um
- College of Oriental Medicine, Institute of Oriental Medicine, Kyung Hee University, 1 Hoegi-Dong, Dongdaemun-Gu, Seoul, 130-701, Republic of Korea
- Acupuncture and Meridian Science Research Center, Kyung Hee University, 1 Hoegi-Dong, Dongdaemun-Gu, Seoul, 130-701, Republic of Korea
| | - Tae-Yong Shin
- College of Pharmacy, Woosuk University, Jeonju, Republic of Korea
| | - Hyung-Min Kim
- College of Oriental Medicine, Institute of Oriental Medicine, Kyung Hee University, 1 Hoegi-Dong, Dongdaemun-Gu, Seoul, 130-701, Republic of Korea
| | - Seung-Heon Hong
- College of Pharmacy, Wonkwang University, Iksan, Jeonbuk, 570-749, Republic of Korea
| |
Collapse
|
21
|
Di R, Huang MT, Ho CT. Anti-inflammatory activities of mogrosides from Momordica grosvenori in murine macrophages and a murine ear edema model. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2011; 59:7474-7481. [PMID: 21631112 DOI: 10.1021/jf201207m] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
Momordica grosvenori (Luo Han Guo), grown primarily in Guangxi province in China, has been traditionally used for thousands of years by the Chinese to make hot drinks for the treatment of sore throat and the removal of phlegm. The natural noncaloric sweetening triterpenoid glycosides (mogrosides) contained in the M. grosvenori fruits are also antioxidative, anticarcinogenic, and helpful in preventing diabetic complications. The aim of this study was to assess the anti-inflammatory properties of mogrosides in both murine macrophage RAW 264.7 cells and a murine ear edema model. The results indicate that mogrosides can inhibit inflammation induced by lipopolysaccharides (LPS) in RAW 264.7 cells by down-regulating the expression of key inflammatory genes iNOS, COX-2, and IL-6 and up-regulating some inflammation protective genes such as PARP1, BCL2l1, TRP53, and MAPK9. Similarly, in the murine ear edema model, 12-O-tetradecanoylphorbol-13-acetate-induced inflammation was inhibited by mogrosides by down-regulating COX-2 and IL-6 and up-regulating PARP1, BCL2l1, TRP53, MAPK9, and PPARδ gene expression. This study shows that the anticancer and antidiabetic effects of M. grosvenori may result in part from its anti-inflammatory activity.
Collapse
Affiliation(s)
- Rong Di
- Department of Plant Biology and Pathology, Rutgers University, New Brunswick, NJ 08901, USA.
| | | | | |
Collapse
|
22
|
Lyle DB, Breger JC, Baeva LF, Shallcross JC, Durfor CN, Wang NS, Langone JJ. Low molecular weight hyaluronic acid effects on murine macrophage nitric oxide production. J Biomed Mater Res A 2010; 94:893-904. [PMID: 20336750 DOI: 10.1002/jbm.a.32760] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Hyaluronic acid (HA) is increasingly used for a number of medical device applications. Since the chemical structure of HA is identical no matter its bacterial or animal origin, it should be the ideal biomaterial. However, short term transient inflammatory reactions are common, while rare long-term adverse events may correlate with subclinical chronic inflammation. Concern has been raised that low molecular weight components or degradation fragments from implanted HA may directly stimulate inflammatory reactions. This study examined a panel of HA molecular weights from the unitary disaccharide up to 1.7 x 10(6) Dalton lengths, in which endotoxin was assayed at a very low level (less than 0.03 EU/mg). The murine cell line RAW 264.7, rat splenocytes, and rat adherent differentiated primary macrophages were assayed for nitric oxide production under a variety of inflammatory conditions plus or minus HA. Under the highest inflammatory states, nitric oxide production was mildly suppressed by HMW-HA while slightly augmented by LMW-HA at mg/mL concentrations. However, at micromolar concentrations fragments below 5000 Daltons, thought to have drug-like qualities, were without effect. These data support the hypothesis that if endotoxin is reduced to an extremely low level, LMW-HA may not directly provoke normal tissue macrophage-mediated inflammatory reactions.
Collapse
Affiliation(s)
- Daniel B Lyle
- Center for Devices and Radiological Health, Office of Science and Engineering Laboratories, FDA, Silver Spring, Maryland 20993-002, USA
| | | | | | | | | | | | | |
Collapse
|
23
|
Mehan S, Meena H, Sharma D, Sankhla R. JNK: A Stress-Activated Protein Kinase Therapeutic Strategies and Involvement in Alzheimer’s and Various Neurodegenerative Abnormalities. J Mol Neurosci 2010; 43:376-90. [DOI: 10.1007/s12031-010-9454-6] [Citation(s) in RCA: 121] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2010] [Accepted: 09/16/2010] [Indexed: 01/26/2023]
|
24
|
Brito MA, Vaz AR, Silva SL, Falcão AS, Fernandes A, Silva RFM, Brites D. N-methyl-aspartate receptor and neuronal nitric oxide synthase activation mediate bilirubin-induced neurotoxicity. Mol Med 2010; 16:372-80. [PMID: 20593111 PMCID: PMC2935951 DOI: 10.2119/molmed.2009.00152] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2009] [Accepted: 06/28/2010] [Indexed: 01/02/2023] Open
Abstract
Hyperbilirubinemia may lead to neurotoxicity and neuronal death. Although the mechanisms of nerve cell damage by unconjugated bilirubin (UCB) appear to involve a disruption of the redox status and excitotoxicity, the contribution of nitric oxide (NO·) and of N-methyl-D-aspartate (NMDA) glutamate receptors is unclear. We investigated the role of NO· and NMDA glutamate receptors in the pathways of nerve cell demise by UCB. Neurons were incubated with 100 micromol/L UCB, in the presence of 100 micromol/L human serum albumin for 4 h at 37ºC, alone or in combination with N-ω-nitro-L-arginine methyl ester (L-NAME) (an inhibitor of neuronal nitric oxide synthase [nNOS]), hemoglobin (an NO· scavenger) or (+)-5-methyl-10,11-dihydro-5H-dibenzo[a,d]cyclohepten-5,10-imine maleate (MK-801) (an NMDA-receptor antagonist). Exposure to UCB led to increased expression of nNOS and production of both NO· and cyclic guanosine 3',5'-monophosphate (cGMP), along with protein oxidation and depletion of glutathione. These events concurred for cell dysfunction and death and were counteracted by L-NAME. Moreover, the UCB-induced loss of neuronal viability was abolished by hemoglobin, whereas the activation of nNOS and production of both NO· and cGMP were counteracted by MK-801, resulting in significant protection from cell dysfunction and death. These results reinforce the involvement of oxidative stress by showing that nerve cell damage by UCB is mediated by NO· and therefore is counteracted by NO· inhibitors or scavengers. Our findings strongly suggest that the activation of nNOS and neurotoxicity occur through the engagement of NMDA receptors. These data reveal a role for overstimulation of glutamate receptors in mediating oxidative damage by UCB.
Collapse
Affiliation(s)
- Maria A Brito
- Research Institute for Medicines and Pharmaceutical Sciences, Faculdade de Farmácia, University of Lisbon, Lisbon, Portugal.
| | | | | | | | | | | | | |
Collapse
|
25
|
Di Paola R, Crisafulli C, Mazzon E, Genovese T, Paterniti I, Bramanti P, Cuzzocrea S. Effect of PD98059, a selective MAPK3/MAPK1 inhibitor, on acute lung injury in mice. Int J Immunopathol Pharmacol 2010; 22:937-50. [PMID: 20074457 DOI: 10.1177/039463200902200409] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
The aim of the present study is to evaluate the contribution of mitogen-activated protein kinase 1-3 MAPK3/MAPK1) in a model of acute lung inflammation in mice. Injection of carrageenan into the pleural cavity of mice elicited an acute inflammatory response characterized by: accumulation of fluid containing a large number of neutrophils (PMNs) in the pleural cavity, infiltration of PMNs in lung tissues and subsequent adhesion molecule expression (I-CAM and P-selectin), lipid peroxidation, and increased production of tumour necrosis factor-alpha, (TNF-alpha) and interleukin-1beta (IL-1beta). Furthermore, carrageenan induced lung apoptosis (Bax and Bcl-2 expression) as well as nitrotyrosine formation, NF-kB activation, and pJNK expression, as determined by immunohistochemical analysis of lung tissues and the degree of lung inflammation and tissue injury (histological score). Administration of PD98059, an inhibitor of MAPK3/MAPK1 (10 mg/kg) 1 h after carrageenan caused a reduction in all the parameters of inflammation measured. Thus, based on these findings we propose that inhibitors of the MAPK3/MAPK1 signaling pathways, such as PD98059, may be useful in the treatment of various inflammatory diseases.
Collapse
Affiliation(s)
- R Di Paola
- IRCCS Centro Neurolesi Bonino-Pulejo, Messina, Italy
| | | | | | | | | | | | | |
Collapse
|
26
|
The role of endogenous and exogenous ligands for the peroxisome proliferator-activated receptor alpha (PPAR-alpha) in the regulation of inflammation in macrophages. Shock 2009; 32:62-73. [PMID: 19533851 DOI: 10.1097/shk.0b013e31818bbad6] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The aim of the present study was to evaluate the role of endogenous and exogenous peroxisome proliferator-activated receptor alpha (PPAR-alpha), a nuclear receptor, on the regulation of inflammation in macrophages. To address this question, we have stimulated peritoneal macrophages from PPAR-alpha wild-type mice and PPAR-alpha knockout mice (PPAR-alpha) with 10 microg/mL LPS and 100 U/mL IFN-gamma. We report here that the absence of a functional PPAR-alpha gene in PPAR-alpha knockout mice resulted in a significant augmentation of various inflammatory parameters in peritoneal macrophages. In particular, we have clearly demonstrated that PPAR-alpha gene deletion increases (1) the mitogen-activated protein kinase phosphorylation (extracellular signal-regulated kinase, c-Jun NH2-terminal kinase, and p38), (2) nuclear factor-kappaB activation, (3) IkappaB-alpha degradation, (4) iNOS expression and NO formation, and (5) cyclooxygenase 2 expression and prostaglandin E2 formation caused by LPS/IFN-gamma stimulation. On the contrary, the incubation of peritoneal macrophages from PPAR-alpha wild type with clofibrate (2 mM) at 2 h before the LPS and IFN-gamma stimulation significantly reduced the expression and the release of the proinflammatory mediators. To elucidate whether the protective effects of clofibrate is related to activation of the PPAR-alpha receptor, we also investigated the effect of clofibrate treatment on PPAR-alpha-deficient mice. The absence of the PPAR-alpha receptor significantly abolished the protective effect of the PPAR-alpha agonist against LPS/IFN-gamma-induced macrophage inflammation. In conclusion, our study demonstrates that the endogenous and exogenous PPAR-alpha ligands reduce the degree of macrophage inflammation caused by LPS/IFN-gamma stimulation.
Collapse
|
27
|
Vause CV, Durham PL. CGRP stimulation of iNOS and NO release from trigeminal ganglion glial cells involves mitogen-activated protein kinase pathways. J Neurochem 2009; 110:811-21. [PMID: 19457095 DOI: 10.1111/j.1471-4159.2009.06154.x] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Clinical and basic science data support an integral role of calcitonin gene-related peptide (CGRP) in the pathophysiology of temporomandibular joint disorders. Recently, we have shown that CGRP can stimulate the synthesis and release of nitric oxide (NO) from trigeminal ganglion glial cells. The goal of this study was to determine the role of mitogen-activated protein kinase (MAPK) signaling pathways in CGRP regulation of iNOS expression and NO release from cultured trigeminal ganglion glial cells from Sprague-Dawley rats. CGRP treatment for 2 h significantly increased activity of the MAPK reporter genes, Elk, ATF-2, and CHOP. In addition, CGRP increased nuclear staining for the active forms of the MAPKs: extracellular signal-regulated kinase, c-Jun amino-terminal kinase, and p38. This stimulatory event was not observed in cultures pre-treated with the CGRP receptor antagonist peptide CGRP(8-37). Similarly, pre-treatment with selective MAPK inhibitors repressed increases in reporter gene activity as well as CGRP-induced increases in iNOS expression and NO release mediated by MAPKs. In addition, over-expression of MAPK kinase 1 (MEK1), MEK3, MEK6, and MEK kinase significantly increased iNOS expression and NO production in glial cells. Results from our study provide evidence that CGRP binding to its receptor can stimulate iNOS gene expression via activation of MAPK pathways in trigeminal ganglion glial cells.
Collapse
Affiliation(s)
- C V Vause
- Department of Biology, Missouri State University, Springfield, Missouri 65897, USA
| | | |
Collapse
|
28
|
Daginakatte GC, Gianino SM, Zhao NW, Parsadanian AS, Gutmann DH. Increased c-Jun-NH2-kinase signaling in neurofibromatosis-1 heterozygous microglia drives microglia activation and promotes optic glioma proliferation. Cancer Res 2009; 68:10358-66. [PMID: 19074905 DOI: 10.1158/0008-5472.can-08-2506] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Neurofibromatosis-1 (NF1) is a common tumor predisposition syndrome in which affected individuals develop benign and malignant tumors. Previous studies from our laboratory and others have shown that benign tumor formation in Nf1 genetically engineered mice (GEM) requires a permissive tumor microenvironment. In the central nervous system, Nf1 loss in glia is insufficient for glioma formation unless coupled with Nf1 heterozygosity in the brain. Our subsequent studies identified Nf1+/- microglia as a critical cellular determinant of optic glioma growth in Nf1 GEM. Using NF1 as an experimental paradigm to further characterize the role of microglia in glioma growth, we first examined the properties of Nf1+/- microglia in vitro and in vivo. Nf1+/- microglia exhibit increased proliferation and motility and express elevated levels of genes associated with microglia activation. We further show that Nf1+/- microglia harbor high levels of activated c-Jun-NH(2)-kinase (JNK) without any significant changes in Akt, mitogen-activated protein kinase (MAPK), or p38-MAPK activity. In contrast, Nf1-/- astrocytes do not exhibit increased JNK activation. SP600125 inhibition of JNK activity in Nf1+/- microglia results in amelioration of the increased proliferation and motility phenotypes and reduces the levels of expression of activated microglia-associated transcripts. Moreover, SP600125 treatment of Nf1 optic glioma-bearing GEM results in reduced optic glioma proliferation in vivo. Collectively, these findings suggest that Nf1+/- microglia represent a good model system to study the role of specialized microglia in brain tumorigenesis and identify a unique Nf1 deregulated pathway for therapeutic studies aimed at abrogating microenvironmental signals that promote brain tumor growth.
Collapse
Affiliation(s)
- Girish C Daginakatte
- Department of Neurology, Washington University School of Medicine, St Louis, Missouri 63110, USA
| | | | | | | | | |
Collapse
|
29
|
Esposito E, Genovese T, Caminiti R, Bramanti P, Meli R, Cuzzocrea S. Melatonin reduces stress-activated/mitogen-activated protein kinases in spinal cord injury. J Pineal Res 2009; 46:79-86. [PMID: 19090911 DOI: 10.1111/j.1600-079x.2008.00633.x] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Permanent functional deficits following spinal cord injury (SCI) arise from both mechanical injury and from secondary tissue reactions involving inflammation. The mitogen-activated protein kinases (MAPKs) play a critical role in cell signaling and gene expression. MAPK family includes three major members: extracellular signal regulated kinase (ERK), p38, and c-Jun N-terminal kinase (JNK), representing three different signaling cascades. Moreover, various studies have clearly shown that high-mobility group box 1 (HMGB1) protein is implicated as a putative danger signal involved in the pathogenesis of a variety of inflammatory conditions including autoimmunity, cancer, trauma and hemorrhagic shock, and ischemia-reperfusion injury. Recently, we have reported that the pineal secretory product melatonin exerts important anti-inflammatory effects in an experimental model of SCI induced by the application of vascular clips (force of 24 g) to the dura after a four-level T5-T8 laminectomy. However, no reports are available on the effect of melatonin on MAPK signaling pathways and HMGB1 expression in SCI. The aim of the present study was to evaluate whether the melatonin protective effect observed in SCI is related to the regulation of MAPK signaling pathways and HMGB1 in mice. In this study we demonstrate the efficacy of treatment with the melatonin in SCI in mice in reducing (a) motor recovery, (b) activation of MAPKs p38, JNK and ERK1/2, (c) tumor necrosis factor-alpha expression, and (d) expression of HMGB1. We propose that melatonin's ability to reduce SCI in mice is also related to a reduction in MAPK signaling pathways and HMGB1 expression.
Collapse
Affiliation(s)
- Emanuela Esposito
- Department of Experimental Pharmacology, University of Naples "Federico II", Napoli, Italy
| | | | | | | | | | | |
Collapse
|
30
|
Martucci C, Trovato AE, Costa B, Borsani E, Franchi S, Magnaghi V, Panerai AE, Rodella LF, Valsecchi AE, Sacerdote P, Colleoni M. The purinergic antagonist PPADS reduces pain related behaviours and interleukin-1β, interleukin-6, iNOS and nNOS overproduction in central and peripheral nervous system after peripheral neuropathy in mice. Pain 2008; 137:81-95. [PMID: 17900807 DOI: 10.1016/j.pain.2007.08.017] [Citation(s) in RCA: 126] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2007] [Revised: 08/06/2007] [Accepted: 08/13/2007] [Indexed: 12/22/2022]
Abstract
Neuropathic pain consequent to peripheral injury is associated with local inflammation and overexpression of nitric oxide synthases (NOS) and inflammatory cytokines in locally recruited macrophages, Schwann and glial cells. We investigated the time course and localization of nitric oxide synthases (NOS) and cytokines in the central (spinal cord and thalamus) and peripheral nervous system (nerve and dorsal root ganglia), in a mouse model of mononeuropathy induced by sciatic nerve chronic constriction injury. ATP is recognized as an endogenous pain mediator. Therefore we also evaluated the role of purinergic signalling in pain hypersensitivity employing the P2 receptor antagonist, pyridoxalphosphate-6-azophenyl-2',4'-disulphonic acid (PPADS), on pain behaviour, NOS and cytokines. The PPADS daily administration starting on day 3 after injury dose- and time-dependently decreased both tactile allodynia and thermal hyperalgesia. PPADS (25mg/kg) completely reversed nociceptive hypersensitivity and simultaneously reduced the increased NO/NOS system and IL-1beta in both peripheral (injured sciatic nerve and L4-L6 ipsilateral dorsal root ganglia) and central steps of nervous system (L4-L6 spinal cord and thalamus) involved in pain signalling. IL-6 was overexpressed only in the peripheral nervous system and PPADS prolonged administration reduced it in sciatic nerve. In conclusion, we hypothesize that NO/NOS and IL-1beta have a pronociceptive role in this neuropathy model, and that purinergic antagonism reduces pain hypersensitivity by inhibiting their overactivity.
Collapse
Affiliation(s)
- Cataldo Martucci
- Department of Pharmacology, Chemotherapy and Medical Toxicology, University of Milano, via Vanvitelli 32, 20129 Milano, Italy Department of Biotechnology and Bioscience, University of Milano-Bicocca, 20126 Milano, Italy Department of Endocrinology and Center of Excellence of Neurodegenerative Diseases, University of Milano, 20133 Milano, Italy Division of Anatomy, Department of Biomedical Sciences and Biotechnology, University of Brescia, Brescia, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Genovese T, Esposito E, Mazzon E, Muià C, Di Paola R, Meli R, Bramanti P, Cuzzocrea S. Evidence for the role of mitogen-activated protein kinase signaling pathways in the development of spinal cord injury. J Pharmacol Exp Ther 2008; 325:100-14. [PMID: 18180375 DOI: 10.1124/jpet.107.131060] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Mitogen-activated protein kinase (MAPK) signaling pathways involve two closely related MAPKs, known as extracellular signal-regulated kinase (ERK)1 and ERK2. The aim of the present study was to evaluate the contribution of MAPK3/MAPK1 in the secondary damage in experimental spinal cord injury (SCI) in mice. To this purpose, we used 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one (PD98059), which is an inhibitor of MAPK3/MAPK1. Spinal cord trauma was induced by the application of vascular clips (force of 24 g) to the dura via a four-level T5-T8 laminectomy. SCI in mice resulted in severe trauma characterized by edema, neutrophil infiltration, and production of inflammatory mediators, tissue damage, and apoptosis. PD98059 treatment (10 mg/kg i.p.) at 1 and 6 h after the SCI significantly reduced 1) the degree of spinal cord inflammation and tissue injury (histological score), 2) neutrophil infiltration (myeloperoxidase activity), 3) nitrotyrosine formation, 4) proinflammatory cytokines expression, 5) nuclear factor-kappaB activation, 6) phospho-ERK1/2 expression, and 6) apoptosis (terminal deoxynucleotidyl transferase dUTP nick-end labeling staining, Fas ligand, Bax, and Bcl-2 expression). Moreover, PD98059 significantly ameliorated the recovery of limb function (evaluated by motor recovery score) in a dose-dependent manner. Taken together, our results clearly demonstrate that PD98059 treatment reduces the development of inflammation and tissue injury associated with spinal cord trauma.
Collapse
Affiliation(s)
- Tiziana Genovese
- Department of Clinical and Experimental Medicine and Pharmacology, School of Medicine, University of Messina, Torre Biologica-Policlinico Universitario Via C. Valeria-Gazzi, 98100 Messina, Italy
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Protective Effects of Aspirin Against Oxidized LDL-induced Inflammatory Protein Expression in Human Endothelial Cells. J Cardiovasc Pharmacol 2008; 51:32-7. [DOI: 10.1097/fjc.0b013e318159ebaf] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
33
|
Stern R, Asari AA, Sugahara KN. Hyaluronan fragments: an information-rich system. Eur J Cell Biol 2006; 85:699-715. [PMID: 16822580 DOI: 10.1016/j.ejcb.2006.05.009] [Citation(s) in RCA: 836] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2006] [Revised: 05/05/2006] [Accepted: 05/05/2006] [Indexed: 02/08/2023] Open
Abstract
Hyaluronan is a straight chain, glycosaminoglycan polymer of the extracellular matrix composed of repeating units of the disaccharide [-D-glucuronic acid-beta1,3-N-acetyl-D-glucosamine-beta1,4-]n. Hyaluronan is synthesized in mammals by at least three synthases with products of varying chain lengths. It has an extraordinary high rate of turnover with polymers being funneled through three catabolic pathways. At the cellular level, it is degraded progressively by a series of enzymatic reactions that generate polymers of decreasing sizes. Despite their exceedingly simple primary structure, hyaluronan fragments have extraordinarily wide-ranging and often opposing biological functions. There are large hyaluronan polymers that are space-filling, anti-angiogenic, immunosuppressive, and that impede differentiation, possibly by suppressing cell-cell interactions, or ligand access to cell surface receptors. Hyaluronan chains, which can reach 2 x 10(4) kDa in size, are involved in ovulation, embryogenesis, protection of epithelial layer integrity, wound repair, and regeneration. Smaller polysaccharide fragments are inflammatory, immuno-stimulatory and angiogenic. They can also compete with larger hyaluronan polymers for receptors. Low-molecular-size polymers appear to function as endogenous "danger signals", while even smaller fragments can ameliorate these effects. Tetrasaccharides, for example, are anti-apoptotic and inducers of heat shock proteins. Various fragments trigger different signal transduction pathways. Particular hyaluronan polysaccharides are also generated by malignant cells in order to co-opt normal cellular functions. How the small hyaluronan fragments are generated is unknown, nor is it established whether the enzymes of hyaluronan synthesis and degradation are involved in maintaining proper polymer sizes and concentration. The vast range of activities of hyaluronan polymers is reviewed here, in order to determine if patterns can be detected that would provide insight into their production and regulation.
Collapse
Affiliation(s)
- Robert Stern
- Department of Pathology and UCSF Comprehensive Cancer Center, School of Medicine, University of California San Francisco, 513 Parnassus Avenue, S-564, San Francisco, CA 94143-0511, USA.
| | | | | |
Collapse
|
34
|
Xu Z, Wang BR, Wang X, Kuang F, Duan XL, Jiao XY, Ju G. ERK1/2 and p38 mitogen-activated protein kinase mediate iNOS-induced spinal neuron degeneration after acute traumatic spinal cord injury. Life Sci 2006; 79:1895-905. [PMID: 16978658 DOI: 10.1016/j.lfs.2006.06.023] [Citation(s) in RCA: 93] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2006] [Revised: 06/10/2006] [Accepted: 06/13/2006] [Indexed: 11/30/2022]
Abstract
The enhanced production of nitric oxide (NO) via inducible nitric oxide synthase (iNOS) has been implicated in the pathogenesis of neuronal apoptosis after acute traumatic spinal cord injury (SCI). In the present study, to further characterize the pathways mediating the synthesis and release of NO, we examined activation of extracellular signal regulated kinase 1/2 (ERK1/2) and p38 mitogen-activated protein kinases (p38 MAPK) in microglia/macrophages in the injured area of adult rats subjected to a complete transection at the T10 vertebrae level and assessed their role in NO production and survival of neurons by using immunohistochemistry, Western blot, RT-PCR and pharmacological interventions. Results showed activation of microglia/macrophages featured by morphological changes, as visualized immunohistochemically with the marker OX-42, in the areas adjacent to the lesion epicenter 1 h after surgery. Concomitantly, iNOS mRNA and its protein in the activated microglia/macrophages were also significantly upregulated at early hours after surgery. Their levels were maximal at 6 h, persisted for at least 24 h, and returned to basal level 72 h after SCI. Furthermore, phosphorylated ERK1/2 and p38 MAPK were activated as well in microglia/macrophages in injured area with a similar time course as iNOS. With administration of L-NAME, a NOS inhibitor, the number of apoptotic neurons was clearly decreased, as assessed with TUNEL method at 24 h after SCI. In parallel, loss of neurons induced by SCI, assessed with NeuN immunohistochemistry, was also diminished. Moreover, the effect of inhibition of phosphorylation ERK1/2 and p38 MAPK by corresponding inhibitors PD98059 and SB203580 administered before and after SCI was also investigated. Inhibition of p38 effectively reduced iNOS mRNA expression and rescued neurons from apoptosis and death in the area adjacent to the lesion epicenter; whereas the inhibition of ERK1/2 had a smaller effect on decrease of iNOS mRNA and no long-term protective effect on cell loss. These results indicate the ERK1/2 and p38 MAPK signaling pathway, especially the latter, play an important role in NO-mediated degeneration of neuron in the spinal cord following SCI. Strategies directed to blocking the initiation of this cascade prove to be beneficial for the treatment of acute SCI.
Collapse
Affiliation(s)
- Zhen Xu
- Institute of Neurosciences, The Fourth Military Medical University, Xi'an, 710032, PR China
| | | | | | | | | | | | | |
Collapse
|
35
|
Abadir PM, Periasamy A, Carey RM, Siragy HM. Angiotensin II type 2 receptor-bradykinin B2 receptor functional heterodimerization. Hypertension 2006; 48:316-22. [PMID: 16754789 DOI: 10.1161/01.hyp.0000228997.88162.a8] [Citation(s) in RCA: 122] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Angiotensin II type 2 (AT2R) or bradykinin B2 (B2R) receptor activation enhances NO production. Recently, we demonstrated enhancement of NO production when AT2R and B2R are simultaneously activated in vivo. However, the mechanism involved in this enhancement is unknown. Using confocal fluorescence resonance energy transfer microscopy, we report the distance between the AT2R and B2R in PC12W cell membranes to be 50+/-5 A, providing evidence and quantification of receptor heterodimerization as the mechanism for enhancing NO production. The rate of AT2R-B2R heterodimer formation is largely a function of the degree of AT2R-B2R expression. The physical association between the dimerized receptors initiates changes in intracellular phosphoprotein signaling activities leading to phosphorylation of c-Jun terminal kinase, phosphotyrosine phosphatase, inhibitory protein kappaBalpha, and activating transcription factor 2; dephosphorylation of p38 and p42/44 mitogen-activated protein kinase and signal transducer inhibitor of transcription 3; and enhancing production of NO and cGMP. Controlling the expression of AT2R-B2R, consequently influencing their biologically active dimerization, presents a potential therapeutic target for the treatment of hypertension and other cardiovascular and renal disorders.
Collapse
Affiliation(s)
- Peter M Abadir
- Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia, Charlottesville, VA, USA
| | | | | | | |
Collapse
|
36
|
Abstract
Elevated levels of NO produced within the central nervous system (CNS) are associated with the pathogenesis of neuroinflammatory and neurodegenerative human diseases such as multiple sclerosis, HIV dementia, brain ischemia, trauma, Parkinson's disease, and Alzheimer's disease. Resident glial cells in the CNS (astroglia and microglia) express inducible nitric oxide synthase (iNOS) and produce high levels of NO in response to a wide variety of proinflammatory and degenerative stimuli. Although pathways resulting in the expression of iNOS may vary in two different glial cells of different species, the intracellular signaling events required for the expression of iNOS in these cells are slowly becoming clear. Various signaling cascades converge to activate several transcription factors that control the transcription of iNOS in glial cells. The present review summarizes different results and discusses current understandings about signaling mechanisms for the induction of iNOS expression in activated glial cells. A complete understanding of the regulation of iNOS expression in glial cells is expected to identify novel targets for therapeutic intervention in NO-mediated neurological disorders.
Collapse
Affiliation(s)
- Ramendra N Saha
- Department of Oral Biology, Section of Neuroscience, University of Nebraska Medical Center, Lincoln, 68583, USA
| | | |
Collapse
|
37
|
Wang MJ, Kuo JS, Lee WW, Huang HY, Chen WF, Lin SZ. Translational event mediates differential production of tumor necrosis factor-α in hyaluronan-stimulated microglia and macrophages. J Neurochem 2006; 97:857-71. [PMID: 16573652 DOI: 10.1111/j.1471-4159.2006.03776.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Recent evidence has demonstrated that hyaluronan synthase 2 mRNA is up-regulated after brain ischemia. After a cerebral ischemic event, microglia and macrophages are the major inflammatory cells and are activated by hyaluronan (HA). However, it is unclear how these cells compare with regard to HA responsiveness. We show here that peritoneal macrophages and RAW 264.7 macrophages produced more than five- and 10-fold more tumor necrosis factor-alpha (TNF-alpha) than primary microglia and BV-2 microglia, respectively. Antibody blockade study showed that CD44, Toll-like receptor-4 receptor and the receptor for HA-mediated motility were responsible for HA-induced TNF-alpha release. Furthermore, HA induced higher levels of phosphorylated MAPK in RAW 264.7 cells when compared with BV-2 cells. HA-mediated TNF-alpha production required p38 MAPK, extracellular-regulated kinase and c-Jun N-terminal kinase phosphorylation in both cell types. The levels of HA-induced TNF-alpha mRNA expression in BV-2 cells were only twofold lower compared with RAW 264.7 cells, suggesting that a translational event is involved in the differential production of TNF-alpha. Western blot analysis revealed that HA treatment resulted in more rapid phosphorylation of eukaryotic initiation factor 4E-binding protein 1 (4E-BP1) and more effective dissociation of 4E-BP1 from eukaryotic initiation factor 4E in RAW 264.7 cells than in BV-2 cells. Additionally, HA-induced phosphorylation of 4E-BP1 was dependent on MAPK signaling, indicating that RAW 264.7 cells exhibited higher levels of hyperphosphorylated 4E-BP1 possibly due to the overactivation of MAPK. The results suggest that resident microglia and blood-derived monocytes/macrophages exhibit differential sensitivities in response to extracellular mediators after brain ischemia.
Collapse
Affiliation(s)
- Mei-Jen Wang
- Neuro-Medical Scientific Center, Buddhist Tzu-Chi General Hospital, Tzu-Chi College of Technology, Hualien, Taiwan
| | | | | | | | | | | |
Collapse
|
38
|
Singleton KD, Beckey VE, Wischmeyer PE. GLUTAMINE PREVENTS ACTIVATION OF NF-kappaB AND STRESS KINASE PATHWAYS, ATTENUATES INFLAMMATORY CYTOKINE RELEASE, AND PREVENTS ACUTE RESPIRATORY DISTRESS SYNDROME (ARDS) FOLLOWING SEPSIS. Shock 2006; 24:583-9. [PMID: 16317391 DOI: 10.1097/01.shk.0000185795.96964.71] [Citation(s) in RCA: 103] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Glutamine (GLN) has been shown to attenuate cytokine release from LPS-stimulated human peripheral blood mononuclear cells; however, the in vivo antiinflammatory effect of GLN in polymicrobial sepsis and ARDS is unknown. This study evaluates the effect of GLN on inflammatory cytokine release and the pathways that may mediate antiinflammatory effects of GLN in the lung. Either 0.75 g/kg of GLN or saline placebo (SP) was administered to male rats 1 h after cecal ligation and puncture (CLP). NF-kappaB activation, IKBalpha degradation, phosphorylation of p38 MAPK, ERK, and MKP-1 expression were evaluated in lung tissue 6 h post-CLP. Lung tissue iNOS and eNOS, TNF-alpha, IL-6, and IL-18 cytokines were assayed. Last, lung histopathology for occurrence of ARDS and survival were examined. GLN given 1 h postsepsis led to inhibition of lung tissue NF-kappaB activation (P < 0.001 vs. SP), attenuated degradation of IKBalpha, and inhibited phosphorylation of p38 MAPK, and ERK, pathways critical for cytokine release. GLN treatment increased MKP-1 peptide expression and significantly attenuated TNF-alpha and IL-6 6 h after CLP. IL-18 was attenuated by GLN at multiple time points post-CLP. Further, GLN abrogated increases in lung iNOS expression and enhanced lung eNOS postsepsis. Finally, GLN prevented the histopathologic appearance of ARDS after sepsis and significantly improved survival. These data reveal that GLN exerts an antiinflammatory effect in sepsis that may be mediated via attenuation of multiple pathways of inflammation such as NF-kappaB, p38 MAPK, ERK, and MKP-1. GLN also showed an inhibition of increases in iNOS expression. The antiinflammatory effect of GLN was associated with attenuation of ARDS and mortality.
Collapse
Affiliation(s)
- Kristen D Singleton
- Department of Anesthesiology, University of Colorado Health Sciences Center, Denver, Colorado 80262, USA.
| | | | | |
Collapse
|
39
|
Liu X, Wu JY, Zhou F, Sun XL, Yao HH, Yang Y, Ding JH, Hu G. The regulation of rotenone-induced inflammatory factor production by ATP-sensitive potassium channel expressed in BV-2 cells. Neurosci Lett 2006; 394:131-5. [PMID: 16257489 DOI: 10.1016/j.neulet.2005.10.018] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2005] [Revised: 09/28/2005] [Accepted: 10/06/2005] [Indexed: 12/21/2022]
Abstract
Our previous studies have demonstrated that activating ATP-sensitive potassium channel (K(ATP) channel), not only improved Parkinsonian behavior and neurochemical symptoms, but also reduced iNOS activity and mRNA levels in striatum and nigra of rotenone rat model of Parkinson's disease (PD). In this study, it was first shown that the subunits of K(ATP) channels are expressed in BV-2 cells, and then it was investigated whether K(ATP) channel was involved in regulating inflammatory factor production from BV-2 cells activated by rotenone. It was found that K(ATP) channel was expressed in BV-2 cell and formed by the combination of Kir 6.1 and SUR 2A/2B. K(ATP) channel openers (KCOs) including pinacidil, diazoxide and iptakalim (Ipt) exerted beneficial effects on rotenone-induced morphological alterations of BV-2 cells, decreased tumor necrosis factor alpha (TNF-alpha) production and the expression and activity of inducible isoform of nitric oxide synthase (iNOS). Either glibenclamide or 5-hydroxydecanoate acid (a selective mitochondrial K(ATP) channel blocker) could abolish the effects of KCOs, suggesting that K(ATP) channels, especially mitochondrial ATP-sensitive potassium channels (mitoK(ATP) channels), played a crucial role in preventing the activation of BV-2 cells, and subsequently the production of a variety of proinflammatory factors. Therefore, activation of K(ATP) channel might be a new therapeutic strategy for treating neuroinflammatory and neurodegenerative disorders.
Collapse
Affiliation(s)
- Xing Liu
- Laboratory of Neuropharmacology, Department of Anatomy, Histology & Pharmacology, Institute of Neurosciences, Nanjing Medical University, Jiangsu, PR China
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Nagai A, Mishima S, Ishida Y, Ishikura H, Harada T, Kobayashi S, Kim SU. Immortalized human microglial cell line: phenotypic expression. J Neurosci Res 2005; 81:342-8. [PMID: 15957187 DOI: 10.1002/jnr.20478] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Microglia are a major neuroglial component of the CNS, playing an important role as resident immunocompetent and phagocytic cells in the CNS in the event of injury and disease. To understand the role of microglia in the CNS in health and diseases, we have recently established an immortalized clonal cell line of human microglia, HMO6, from human embryonic telencephalon tissue by using a retroviral vector encoding v-myc. This immortalized microglia HMO6 cell line exhibits cell-type-specific antigens for microglia, including CD11b (Mac-1), CD68, CD86 (B7-2), HLA-ABC, HLA-DR, and RCA-1 lectin, and actively phagocytoses latex beads.
Collapse
Affiliation(s)
- Atsushi Nagai
- Department of Neurology, Shimane University School of Medicine, Izumo, Japan
| | | | | | | | | | | | | |
Collapse
|
41
|
Iacob S, Knudson CB. Hyaluronan fragments activate nitric oxide synthase and the production of nitric oxide by articular chondrocytes. Int J Biochem Cell Biol 2005; 38:123-33. [PMID: 16181799 PMCID: PMC3139231 DOI: 10.1016/j.biocel.2005.08.011] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2005] [Revised: 07/08/2005] [Accepted: 08/18/2005] [Indexed: 01/28/2023]
Abstract
Chondrocyte CD44 receptors anchor hyaluronan to the cell surface, enabling the assembly and retention of proteoglycan aggregates in the pericellular matrix. Hyaluronan-CD44 interactions also provide signaling important for maintaining cartilage homeostasis. Disruption of chondrocyte-hyaluronan contact alters CD44 occupancy, initiating alternative signaling cascades. Treatment with hyaluronan oligosaccharides is one approach to uncouple CD44 receptors from its native ligand, hyaluronan. In bovine articular chondrocytes, treatment with hyaluronan oligosaccharides or purified hyaluronan hexasaccharides induced the production of nitric oxide that mirrored nitric oxide production following interleukin-1 treatment. In contrast, 120 and 1,260 kDa hyaluronan did not induce production of nitric oxide. Human chondrocytes responded similarly to treatment with hyaluronan or hyaluronan oligosaccharides. Nitric oxide production from chondrocytes was mediated by activation of the inducible nitric oxide synthase, as confirmed by mRNA expression and inhibition of nitric oxide production by diphenyleneiodonium. Co-treatment of chondrocytes with hyaluronan oligosaccharides and interleukin-1 did not demonstrate additive effects. Blocking interleukin-1 receptors with an antagonist did not abolish the production of nitric oxide induced by treatment with hyaluronan oligosaccharides. Moreover, only COS-7 following transfection with a pCD44, not the CD44-null parental cells, responded to treatment with hyaluronan oligosaccharides by releasing nitric oxide. This study demonstrates a novel signaling potential by hyaluronan fragments, in lieu of endogenous hyaluronan-chondrocyte interactions, resulting in the activation of inducible nitric oxide synthase.
Collapse
Affiliation(s)
| | - Cheryl B. Knudson
- Corresponding author. Tel.: +1 312 942 8249; fax: +1 312 942 3053., (C.B. Knudson)
| |
Collapse
|
42
|
Back SA, Tuohy TMF, Chen H, Wallingford N, Craig A, Struve J, Luo NL, Banine F, Liu Y, Chang A, Trapp BD, Bebo BF, Rao MS, Sherman LS. Hyaluronan accumulates in demyelinated lesions and inhibits oligodendrocyte progenitor maturation. Nat Med 2005; 11:966-72. [PMID: 16086023 DOI: 10.1038/nm1279] [Citation(s) in RCA: 469] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2005] [Accepted: 07/05/2005] [Indexed: 11/09/2022]
Abstract
Demyelination is the hallmark of numerous neurodegenerative conditions, including multiple sclerosis. Oligodendrocyte progenitors (OPCs), which normally mature into myelin-forming oligodendrocytes, are typically present around demyelinated lesions but do not remyelinate affected axons. Here, we find that the glycosaminoglycan hyaluronan accumulates in demyelinated lesions from individuals with multiple sclerosis and in mice with experimental autoimmune encephalomyelitis. A high molecular weight (HMW) form of hyaluronan synthesized by astrocytes accumulates in chronic demyelinated lesions. This form of hyaluronan inhibits remyelination after lysolecithin-induced white matter demyelination. OPCs accrue and do not mature into myelin-forming cells in demyelinating lesions where HMW hyaluronan is present. Furthermore, the addition of HMW hyaluronan to OPC cultures reversibly inhibits progenitor-cell maturation, whereas degrading hyaluronan in astrocyte-OPC cocultures promotes oligodendrocyte maturation. HMW hyaluronan may therefore contribute substantially to remyelination failure by preventing the maturation of OPCs that are recruited to demyelinating lesions.
Collapse
Affiliation(s)
- Stephen A Back
- Department of Pediatrics, School of Medicine, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, Oregon 97239, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Abstract
Microglia participate in all phases of the multiple sclerosis (MS) disease process. As members of the innate immune system, these cells have evolved to respond to stranger/danger signals; such a response within the central nervous system (CNS) environment has the potential to induce an acute inflammatory response. Engagement of Toll-like receptors (TLRs), a major family of pattern-recognition receptors (PRRs), provides an important mechanism whereby microglia can interact with both exogenous and endogenous ligands within the CNS. Such interactions modulate the capacity of microglia to present antigens to cells of the adaptive immune system and thus contribute to the initiation and propagation of the more sophisticated antigen-directed responses. This inflammatory response introduces the potential for bidirectional feedback between CNS resident and infiltrating systemic cells. Such interactions acquire particular relevance in the era of therapeutics for MS because the infiltrating cells can be subjected to systemic immunomodulatory therapies known to change their functional properties. Phagocytosis by microglia/macrophages is a hallmark of the MS lesion; however, the extent of tissue damage and the type of cell death will dictate subsequent innate responses. Microglia/macrophages are armed with a battery of effector molecules, such as reactive nitrogen species, that may contribute to CNS tissue injury, specifically to the injury of oligodendrocytes that is associated with MS. A therapeutic challenge is to modulate the dynamic properties of microglia/macrophages so as to limit potentially damaging innate responses, to protect the CNS from injury, and to promote local recovery.
Collapse
Affiliation(s)
- Carolyn Jack
- Neuroimmunology Unit, Montreal Neurological Institute, Montreal, Quebec, Canada
| | | | | | | |
Collapse
|
44
|
Kim SJ, Jeong HJ, Choi IY, Lee KM, Park RK, Hong SH, Kim HM. Cyclooxygenase-2 inhibitor SC-236 [4-[5-(4-chlorophenyl)-3-(trifluoromethyl)-1-pyrazol-1-l] benzenesulfonamide] suppresses nuclear factor-kappaB activation and phosphorylation of p38 mitogen-activated protein kinase, extracellular signal-regulated kinase, and c-Jun N-terminal kinase in human mast cell line cells. J Pharmacol Exp Ther 2005; 314:27-34. [PMID: 15784648 DOI: 10.1124/jpet.104.082792] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
SC-236 [4-[5-(4-chlorophenyl)-3-(trifluoromethyl)-1-pyrazol-1-l] benzenesulfonamide; C16H11ClF3N3O2S] is a highly selective cyclooxygenase (COX)-2 inhibitor. However, the exact mechanism that accounts for the anti-inflammatory effect of SC-236 is not completely understood. The aim of the present study was to elucidate whether and how SC-236 modulates the inflammatory reaction in a stimulated human mast cell (HMC) line, HMC-1. SC-236 inhibited the expression of tumor necrosis factor-alpha, interleukin (IL)-6, IL-8, vascular endothelial growth factor, COX-2, inducible nitric-oxide synthase, and hypoxia-inducible factor-1alpha in phorbol 12-myristate 13-acetate plus calcium ionophore A23187 (PMACI)-stimulated HMC-1. SC-236 suppressed nuclear factor (NF)-kappaB activation induced by PMACI, leading to suppression of IkappaB-alpha phosphorylation and degradation. SC-236 also suppressed strong induction of NF-kappaB promoter-mediated luciferase activity. In addition, SC-236 suppressed PMACI-induced phosphorylation of the mitogen-activated protein kinase p38, the extracellular-regulated kinase p44, and the c-Jun N-terminal kinase and induced expression of mitogen-activated protein kinase phosphatase-1. These results provide new insight into the pharmacological actions of SC-236 as a potential molecule for therapy of mast cell-mediated inflammatory diseases.
Collapse
Affiliation(s)
- Su-Jin Kim
- College of Oriental Medicine, Kyung Hee University, Dongdaemun-Gu, Seoul, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
45
|
Werber S, Shalit I, Fabian I, Steuer G, Weiss T, Blau H. Moxifloxacin inhibits cytokine-induced MAP kinase and NF-kappaB activation as well as nitric oxide synthesis in a human respiratory epithelial cell line. J Antimicrob Chemother 2005; 55:293-300. [PMID: 15659543 DOI: 10.1093/jac/dkh525] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND We previously demonstrated that the quinolone moxifloxacin prevents Candida albicans pneumonitis and epithelial nuclear factor kappaB (NF-kappaB) nuclear translocation in immunosuppressed mice. OBJECTIVES To explore the anti-inflammatory effects of moxifloxacin directly on a lung epithelial cell line. METHODS We studied the effect of clinically relevant concentrations of moxifloxacin (2.5-10 mg/L) on cytokine-induced activation of nitric oxide (NO) secretion, inducible NO synthase (iNOS) expression and the activation of signal transduction pathways of inflammation, NF-kappaB and the mitogen-activated protein kinases [extracellular signal-regulated kinases (ERK1/2) and C-Jun N-terminal kinase (JNK)], in the A549 lung epithelial cell line. RESULTS Stimulation with the cytokines interleukin-1beta(IL-1beta)/interferon-gamma (IFN-gamma) increased NO up to 3.3-fold and moxifloxacin inhibited this up to 68% (P < 0.05). Similarly, the increase in iNOS levels was inhibited in cells pre-treated with moxifloxacin by up to 62%. IL-1beta stimulated a rapid increase in the activities of early intracellular signalling molecules, ERK1/2 and JNK. Moxifloxacin inhibited ERK1/2 by up to 100% and p-JNK activation by 100%. NF-kappaB, as measured by electrophoretic mobility shift assay, was inhibited up to 72% by moxifloxacin. Western-blot analysis revealed that IL-1beta enhanced NF-kappaB p65 and p50 proteins by 1.7- and 3.6-fold, respectively, whereas moxifloxacin inhibited the proteins by up to 60%. CONCLUSIONS Moxifloxacin inhibits intracellular signalling, iNOS expression and NO secretion in a lung epithelial cell line. Future studies may uncover a primary site of quinolone immunomodulation, either upstream or at the cell membrane. Eventually, this quinolone might become an important therapy for inflammatory lung diseases.
Collapse
Affiliation(s)
- Sara Werber
- Department of Cell Biology and Histology, Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | | | | | | | | | | |
Collapse
|
46
|
Abstract
Whether or not T cell receptor engagement leads to full activation or tolerance is determined by the context in which the antigen is encountered. Antigen presented by activated APCs in the presence of costimulation leads to full T cell activation, while antigen presented by resting APCs leads to tolerance. Pathogen-associated molecular patterns in the form of toll-like receptor ligands play a critical role in activating APCs and promoting T cell activation. In this review we hypothesize that low-molecular-weight species of the extracellular matrix polymer hyaluronan also performs this function by acting as an endogenous danger signal.
Collapse
Affiliation(s)
- Jonathan D Powell
- Bunting-Blaustein Cancer Research Building, Johns Hopkins University School of Medicine, 1650 Orleans Street, Baltimore, MD 21231, USA.
| | | |
Collapse
|