1
|
Sharma V, Sharma P, Singh TG. Leukotriene signaling in neurodegeneration: implications for treatment strategies. Inflammopharmacology 2024; 32:3571-3584. [PMID: 39167313 DOI: 10.1007/s10787-024-01557-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 08/11/2024] [Indexed: 08/23/2024]
Abstract
Leukotrienes (LTs) are a group of substances that cause inflammation. They are produced by the enzyme 5-lipoxygenase (5-LOX) from arachidonic acid. Cysteinyl LTs are a group of lipid molecules that have a prominent role in inflammatory signaling in the allergic diseases. Although they are traditionally known for their role in allergic disease, current advancements in bio-medical research have shed light on the involvement of these inflammatory mediators in diseases such as in the inflammation related to central nervous system (CNS) disorders. Among the CNS diseases, LTs, along with 5-LOX and their receptors, have been shown to be associated with multiple sclerosis (MS), Alzheimer's disease (AD), and Parkinson's disease (PD). Through a comprehensive review of current research and experimentation, this investigation provides an insight on the biosynthesis, receptors, and biological effects of LTs in the body. Furthermore, implications of leukotriene signaling in CNS and its intricate role in neurodegeneration are also studied. Through the revelation of these insights, our aim is to establish a foundation for the development of enhanced and focused therapeutic approaches in the continuous endeavor to combat neurodegeneration. Furthermore, the pharmacological inhibition of leukotriene signaling with selective inhibitors offers promising prospects for future interventions and treatments for neurodegenerative diseases.
Collapse
Affiliation(s)
- Veerta Sharma
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India
| | - Prateek Sharma
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India
| | - Thakur Gurjeet Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India.
| |
Collapse
|
2
|
Sood R, Anoopkumar-Dukie S, Rudrawar S, Hall S. Neuromodulatory effects of leukotriene receptor antagonists: A comprehensive review. Eur J Pharmacol 2024; 978:176755. [PMID: 38909933 DOI: 10.1016/j.ejphar.2024.176755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 06/09/2024] [Accepted: 06/16/2024] [Indexed: 06/25/2024]
Abstract
Cysteinyl leukotrienes (CysLTs) are central to the pathophysiology of asthma and various inflammatory disorders. Leukotriene receptor antagonists (LTRAs) effectively treat respiratory conditions by targeting cysteinyl leukotriene receptors, CysLT1 and CysLT2 subtypes. This review explores the multifaceted effects of LTs, extending beyond bronchoconstriction. CysLT receptors are not only present in the respiratory system but are also crucial in neuronal signaling pathways. LTRAs modulate these receptors, influencing downstream signaling, calcium levels, inflammation, and oxidative stress (OS) within neurons hinting at broader implications. Recent studies identify novel molecular targets, sparking interest in repurposing LTRAs for therapeutic use. Clinical trials are investigating their potential in neuroinflammation control, particularly in Alzheimer's disease (AD) and Parkinson's diseases (PD). However, montelukast, a long-standing LTRA since 1998, raises concerns due to neuropsychiatric adverse drug reactions (ADRs). Despite widespread use, understanding montelukast's metabolism and underlying ADR mechanisms remains limited. This review comprehensively examines LTRAs' diverse biological effects, emphasizing non-bronchoconstrictive activities. It also analyses plausible mechanisms behind LTRAs' neuronal effects, offering insights into their potential as neurodegenerative disease modulators. The aim is to inform clinicians, researchers, and pharmaceutical developers about LTRAs' expanding roles, particularly in neuroinflammation control and their promising repurposing for neurodegenerative disease management.
Collapse
Affiliation(s)
- Radhika Sood
- School of Pharmacy and Medical Sciences, Griffith University, Queensland, 4222, Australia
| | | | - Santosh Rudrawar
- School of Pharmacy and Medical Sciences, Griffith University, Queensland, 4222, Australia; Institute for Glycomics, Griffith University, Queensland, 4222, Australia
| | - Susan Hall
- School of Pharmacy and Medical Sciences, Griffith University, Queensland, 4222, Australia.
| |
Collapse
|
3
|
Yuan NY, Medders KE, Sanchez AB, Shah R, de Rozieres CM, Ojeda-Juárez D, Maung R, Williams R, Gelman BB, Baaten BJ, Roberts AJ, Kaul M. A critical role for Macrophage-derived Cysteinyl-Leukotrienes in HIV-1 induced neuronal injury. Brain Behav Immun 2024; 118:149-166. [PMID: 38423397 PMCID: PMC11173376 DOI: 10.1016/j.bbi.2024.02.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 01/26/2024] [Accepted: 02/21/2024] [Indexed: 03/02/2024] Open
Abstract
Macrophages (MΦ) infected with human immunodeficiency virus (HIV)-1 or activated by its envelope protein gp120 exert neurotoxicity. We found previously that signaling via p38 mitogen-activated protein kinase (p38 MAPK) is essential to the neurotoxicity of HIVgp120-stimulated MΦ. However, the associated downstream pathways remained elusive. Here we show that cysteinyl-leukotrienes (CysLT) released by HIV-infected or HIVgp120 stimulated MΦ downstream of p38 MAPK critically contribute to neurotoxicity. SiRNA-mediated or pharmacological inhibition of p38 MAPK deprives MΦ of CysLT synthase (LTC4S) and, pharmacological inhibition of the cysteinyl-leukotriene receptor 1 (CYSLTR1) protects cerebrocortical neurons against toxicity of both gp120-stimulated and HIV-infected MΦ. Components of the CysLT pathway are differentially regulated in brains of HIV-infected individuals and a transgenic mouse model of NeuroHIV (HIVgp120tg). Moreover, genetic ablation of LTC4S or CysLTR1 prevents neuronal damage and impairment of spatial memory in HIVgp120tg mice. Altogether, our findings suggest a novel critical role for cysteinyl-leukotrienes in HIV-associated brain injury.
Collapse
Affiliation(s)
- Nina Y Yuan
- University of California Riverside, School of Medicine, Division of Biomedical Sciences, 900 University Ave, Riverside, CA 92521, USA.
| | - Kathryn E Medders
- Sanford Burnham Prebys Medical Discovery Institute, Infectious and Inflammatory Disease Center, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA.
| | - Ana B Sanchez
- Sanford Burnham Prebys Medical Discovery Institute, Infectious and Inflammatory Disease Center, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA.
| | - Rohan Shah
- University of California Riverside, School of Medicine, Division of Biomedical Sciences, 900 University Ave, Riverside, CA 92521, USA.
| | - Cyrus M de Rozieres
- Sanford Burnham Prebys Medical Discovery Institute, Infectious and Inflammatory Disease Center, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA.
| | - Daniel Ojeda-Juárez
- University of California Riverside, School of Medicine, Division of Biomedical Sciences, 900 University Ave, Riverside, CA 92521, USA; Sanford Burnham Prebys Medical Discovery Institute, Infectious and Inflammatory Disease Center, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA.
| | - Ricky Maung
- University of California Riverside, School of Medicine, Division of Biomedical Sciences, 900 University Ave, Riverside, CA 92521, USA; Sanford Burnham Prebys Medical Discovery Institute, Infectious and Inflammatory Disease Center, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA.
| | - Roy Williams
- Sanford Burnham Prebys Medical Discovery Institute, Infectious and Inflammatory Disease Center, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA.
| | - Benjamin B Gelman
- Department of Pathology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555-0419 USA; Department of Neurobiology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555-0419 USA.
| | - Bas J Baaten
- Sanford Burnham Prebys Medical Discovery Institute, Infectious and Inflammatory Disease Center, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA.
| | - Amanda J Roberts
- Animal Models Core, The Scripps Research Institute, La Jolla, CA 92037, USA.
| | - Marcus Kaul
- University of California Riverside, School of Medicine, Division of Biomedical Sciences, 900 University Ave, Riverside, CA 92521, USA; Sanford Burnham Prebys Medical Discovery Institute, Infectious and Inflammatory Disease Center, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA.
| |
Collapse
|
4
|
Marques CF, Marques MM, Justino GC. Leukotrienes vs. Montelukast—Activity, Metabolism, and Toxicity Hints for Repurposing. Pharmaceuticals (Basel) 2022; 15:ph15091039. [PMID: 36145259 PMCID: PMC9505853 DOI: 10.3390/ph15091039] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/17/2022] [Accepted: 08/19/2022] [Indexed: 11/16/2022] Open
Abstract
Increasing environmental distress is associated with a growing asthma incidence; no treatments are available but montelukast (MTK)—an antagonist of the cysteinyl leukotrienes receptor 1—is widely used in the management of symptoms among adults and children. Recently, new molecular targets have been identified and MTK has been proposed for repurposing in other therapeutic applications, with several ongoing clinical trials. The proposed applications include neuroinflammation control, which could be explored in some neurodegenerative disorders, such as Alzheimer’s and Parkinson’s diseases (AD and PD). However, this drug has been associated with an increasing number of reported neuropsychiatric adverse drug reactions (ADRs). Besides, and despite being on the market since 1998, MTK metabolism is still poorly understood and the mechanisms underlying neuropsychiatric ADRs remain unknown. We review the role of MTK as a modulator of leukotriene pathways and systematize the current knowledge about MTK metabolism. Known toxic effects of MTK are discussed, and repurposing applications are presented comprehensively, with a focus on AD and PD.
Collapse
Affiliation(s)
- Cátia F. Marques
- Centro de Química Estrutural, Institute of Molecular Sciences, Instituto Superior Técnico, Universidade de Lisboa, 1049-001 Lisboa, Portugal
| | - Maria Matilde Marques
- Centro de Química Estrutural, Institute of Molecular Sciences, Instituto Superior Técnico, Universidade de Lisboa, 1049-001 Lisboa, Portugal
- Departamento de Engenharia Química, Instituto Superior Técnico, Universidade de Lisboa, 1049-001 Lisboa, Portugal
| | - Gonçalo C. Justino
- Centro de Química Estrutural, Institute of Molecular Sciences, Instituto Superior Técnico, Universidade de Lisboa, 1049-001 Lisboa, Portugal
- Correspondence:
| |
Collapse
|
5
|
Yu S, Chen X, Li X, Yan J, Jiang Y. Neuroprotective effects of CysLTR antagonist on Streptococcus pneumoniae‑induced meningitis in rats. Exp Ther Med 2022; 24:443. [PMID: 35720636 PMCID: PMC9185808 DOI: 10.3892/etm.2022.11370] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 04/26/2022] [Indexed: 11/17/2022] Open
Abstract
Cysteinyl leukotrienes (CysLTs) modulate central nervous system inflammatory responses via their receptors, CysLT1R and CysLT2R. It has been demonstrated that CysLTR participates in the infection process of Streptococcus pneumoniae (SP)-induced meningitis. In the present study, the effects and possible underlying mechanisms of CysLTR antagonists (pranlukast and HAMI 3379) on SP meningitis were further determined. SP meningitis was induced by intracerebroventricular injection of serotype III SP in Sprague-Dawley rats which were administrated intraperitoneally with 0.1 mg/kg antagonists. The clinical disease status of rats was evaluated by body weight and behavioral changes with neurological scoring. Survival neuron density, activated microglial and astrocytes were assessed by Nissl staining and immunohistochemical staining. The expression levels of inflammatory cytokines and NLRP3 inflammasome were detected by reverse transcription-quantitative PCR and western blotting, respectively. Pranlukast and HAMI 3379 treatment markedly alleviated the clinical disease status, which was manifested by improving body weight loss and neurological deficit. Furthermore, pranlukast and HAMI 3379 treatment ameliorated neuronal injury and inhibited microgliosis and astrogliosis. In addition, significant downregulation of inflammatory cytokines and NLRP3 expression was observed in pranlukast and HAMI 3379-treated rats. These in vivo findings indicated the neuroprotective effects of CysLTR antagonists against experimental SP-induced meningitis, and the mechanism of anti-inflammatory effects may partly be by inhibiting NLRP3 inflammasome overactivation.
Collapse
Affiliation(s)
- Shuying Yu
- Department of Pharmacy, Hangzhou Children's Hospital, Hangzhou, Zhejiang 310014, P.R. China
| | - Xiaojin Chen
- Department of Pharmacy, Hangzhou Children's Hospital, Hangzhou, Zhejiang 310014, P.R. China
| | - Xiaoyu Li
- Department of Pharmacy, Hangzhou Children's Hospital, Hangzhou, Zhejiang 310014, P.R. China
| | - Jun Yan
- Department of Pharmacy, Hangzhou Children's Hospital, Hangzhou, Zhejiang 310014, P.R. China
| | - Yingying Jiang
- Department of Pharmacy, Hangzhou Children's Hospital, Hangzhou, Zhejiang 310014, P.R. China
| |
Collapse
|
6
|
Leukotriene Signaling as a Target in α-Synucleinopathies. Biomolecules 2022; 12:biom12030346. [PMID: 35327537 PMCID: PMC8944962 DOI: 10.3390/biom12030346] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 01/12/2022] [Accepted: 02/12/2022] [Indexed: 01/04/2023] Open
Abstract
Parkinson’s disease (PD) and dementia with Lewy bodies (DLB) are two common types of α-synucleinopathies and represent a high unmet medical need. Despite diverging clinical manifestations, both neurodegenerative diseases share several facets of their complex pathophysiology. Apart from α-synuclein aggregation, an impairment of mitochondrial functions, defective protein clearance systems and excessive inflammatory responses are consistently observed in the brains of PD as well as DLB patients. Leukotrienes are lipid mediators of inflammatory signaling traditionally known for their role in asthma. However, recent research advances highlight a possible contribution of leukotrienes, along with their rate-limiting synthesis enzyme 5-lipoxygenase, in the pathogenesis of central nervous system disorders. This review provides an overview of in vitro as well as in vivo studies, in summary suggesting that dysregulated leukotriene signaling is involved in the pathological processes underlying PD and DLB. In addition, we discuss how the leukotriene signaling pathway could serve as a future drug target for the therapy of PD and DLB.
Collapse
|
7
|
Potential Effects of Leukotriene Receptor Antagonist Montelukast in Treatment of Neuroinflammation in Parkinson's Disease. Int J Mol Sci 2021; 22:ijms22115606. [PMID: 34070609 PMCID: PMC8198163 DOI: 10.3390/ijms22115606] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 05/16/2021] [Accepted: 05/21/2021] [Indexed: 12/11/2022] Open
Abstract
Parkinson’s disease (PD) is a neurodegenerative disorder where misfolded alpha-synuclein-enriched aggregates called Lewy bodies are central in pathogenesis. No neuroprotective or disease-modifying treatments are currently available. Parkinson’s disease is considered a multifactorial disease and evidence from multiple patient studies and animal models has shown a significant immune component during the course of the disease, highlighting immunomodulation as a potential treatment strategy. The immune changes occur centrally, involving microglia and astrocytes but also peripherally with changes to the innate and adaptive immune system. Here, we review current understanding of different components of the PD immune response with a particular emphasis on the leukotriene pathway. We will also describe evidence of montelukast, a leukotriene receptor antagonist, as a possible anti-inflammatory treatment for PD.
Collapse
|
8
|
Zhao Y, Gan Y, Xu G, Hua K, Liu D. Exosomes from MSCs overexpressing microRNA-223-3p attenuate cerebral ischemia through inhibiting microglial M1 polarization mediated inflammation. Life Sci 2020; 260:118403. [PMID: 32926923 DOI: 10.1016/j.lfs.2020.118403] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 08/30/2020] [Accepted: 09/05/2020] [Indexed: 02/06/2023]
Abstract
AIMS To explore the therapeutic effect and possible mechanism of exosomes from MSCs overexpressing miR-223 on cerebral ischemia and microglia polarization mediated inflammation. MAIN METHODS Rats after middle cerebral artery occlusion and reperfusion (MCAO/R) surgery and microglia BV-2 exposed to oxygen and glucose deprivation (OGD) and cysteinyl leukotrienes (CysLTs) stimulation were subject to exosomes from miR-223-3p transfected MSCs treatment, respectively. Behavioral tests were applied to assess the rats' neurological function. FACS was used to analyze M1/M2 microglia BV-2. production of cytokines in the ischemic hemisphere and BV-2 was detected by ELISA or qRT-PCR. Western blotting and qRT-PCR were also used to examine the expression of cysteinyl leukotriene receptor 2 (CysLT2R) in vivo and in vitro. KEY FINDINGS Exosomes from MSCs over expressing miR-223-3p decreased MCAO/R induced cerebral infarct volume, improved neurological deficits, promoted learning and memorizing abilities. They suppressed pro-inflammatory factors expression and promoted anti-inflammatory factors secretion in the ischemic cortex and hippocampus. In vitro, exosomal miR-223-3p exhibited a more evident impact on modulating mRNA expression and protein production of cytokines. It promoted M2 microglia transformation of M1 microglia induced by NMLTC4 with a concentration-dependent manner. Western blot and qRT-PCR also revealed exosomal miR-223-3p decreased mRNA and protein expression of CysLT2R in vitro and in vivo. SIGNIFICANCE Exosomal miR-223-3p from MSCs attenuated cerebral ischemia/reperfusion injury through inhibiting microglial M1 polarization mediated pro-inflammatory response, which may be related with inhibitory effect of exosomal miR-223-3p on CysLT2R.
Collapse
Affiliation(s)
- Yangmin Zhao
- School of Clinical Sciences, Hangzhou Medical College, Zhejiang, China
| | - Yunxiao Gan
- School of Pharmaceutical Sciences, Hangzhou Medical College, Zhejiang, China
| | - Gewei Xu
- School of Clinical Sciences, Hangzhou Medical College, Zhejiang, China
| | - Kouzhen Hua
- School of Basic Medical Sciences & Forensic Medicine, Hangzhou Medical College, Zhejiang, China
| | - Dandan Liu
- School of Basic Medical Sciences & Forensic Medicine, Hangzhou Medical College, Zhejiang, China.
| |
Collapse
|
9
|
Zhao Y, Gan Y, Xu G, Yin G, Liu D. MSCs-Derived Exosomes Attenuate Acute Brain Injury and Inhibit Microglial Inflammation by Reversing CysLT2R-ERK1/2 Mediated Microglia M1 Polarization. Neurochem Res 2020; 45:1180-1190. [PMID: 32112178 DOI: 10.1007/s11064-020-02998-0] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 02/15/2020] [Accepted: 02/24/2020] [Indexed: 12/12/2022]
Abstract
Inflammatory responses play a major role in the pathophysiology of cerebral ischemia. Mesenchymal stem cell-derived exosomes (MSC-exos) have important anti-inflammatory effects on the treatment of organ injury. This study aimed to determine the anti-inflammatory effect and furtherly investigate the potential mechanism of MSC-exos on acute cerebral ischemia. MSC-exos were isolated by ultracentrifugation, characterized by transmission electron microscopy and FACS. Rats subjected to middle cerebral artery occlusion/reperfusion (MCAO/R) surgery were administered MSC-exos through the tail vein. In vitro, microglia exposed to oxygen- and glucose-deprivation (OGD) and leukotrienes were used to study the protective mechanism of exosomes against ischemia/reperfusion-induced inflammation. The intake of exosomes into microglia was visualized through immunofluorescence staining. The results showed that MSC-exos treatment significantly improved motor, learning and memory abilities of MCAO/R rats 7 days later. The production of pro-inflammatory factors decreased, while the anti-inflammatory cytokines and neurotrophic factors increased both in the cortex and hippocampus of ischemic hemisphere as well as in the culture supernatant of microglia treated with OGD and NMLTC4. MSC-exos treatment also significantly inhibited M1 microglia polarization and increased M2 microglia cells. Furthermore, western blot analysis demonstrated that CysLT2R expression and ERK1/2 phosphorylation were downregulated both in vivo and in vitro. Thus, MSC-exos attenuated brain injury and inhibited microglial inflammation by reversing CysLT2R-ERK1/2 mediated microglia M1 polarization.
Collapse
Affiliation(s)
- Yangmin Zhao
- School of Clinical Sciences, Hangzhou Medical College, Zhejiang, China
| | - Yunxiao Gan
- School of Pharmaceutical Sciences, Hangzhou Medical College, Zhejiang, China
| | - Gewei Xu
- School of Clinical Sciences, Hangzhou Medical College, Zhejiang, China
| | - Guoli Yin
- Shcool of Basic Medical Sciences & Forensic Medicine, Hangzhou Medical College, Zhejiang, China
| | - Dandan Liu
- Shcool of Basic Medical Sciences & Forensic Medicine, Hangzhou Medical College, Zhejiang, China.
| |
Collapse
|
10
|
Modulation of neuroinflammation by cysteinyl leukotriene 1 and 2 receptors: implications for cerebral ischemia and neurodegenerative diseases. Neurobiol Aging 2019; 87:1-10. [PMID: 31986345 DOI: 10.1016/j.neurobiolaging.2019.12.013] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 12/04/2019] [Accepted: 12/14/2019] [Indexed: 12/21/2022]
Abstract
Neuroinflammation is a complex biological process and has been known to play an important role in age-related cerebrovascular and neurodegenerative disorders, such as cerebral ischemia, Alzheimer's disease, and Parkinson's disease. Cysteinyl leukotrienes (CysLTs) are potent inflammatory lipid mediators that exhibit actions mainly through activating type 1 and type 2 CysLT receptors (CysLT1 and CysLT2). Accumulating evidence shows that CysLT1 and CysLT2 are activated at different stages of pathological process in various cell types in the brain such as vascular endothelial cells, astrocytes, microglia, and neurons in response to insults. However, the precise roles and mechanisms of CysLT1 and CysLT2 in regulating the pathogenesis of cerebral ischemia, Alzheimer's disease, and Parkinson's disease are not fully understood. In this article, we focus on current advances that link activation of CysLT1 and CysLT2 to the pathological process during brain ischemia and neurodegeneration and discuss mechanisms by which CysLT1 and CysLT2 mediate inflammatory process and brain injury. Multitarget anti-inflammatory potentials of CysLT1 and CysLT2 antagonism for neuroinflammation and brain injury will also be reviewed.
Collapse
|
11
|
Zhao R, Ying M, Gu S, Yin W, Li Y, Yuan H, Fang S, Li M. Cysteinyl Leukotriene Receptor 2 is Involved in Inflammation and Neuronal Damage by Mediating Microglia M1/M2 Polarization through NF-κB Pathway. Neuroscience 2019; 422:99-118. [PMID: 31726033 DOI: 10.1016/j.neuroscience.2019.10.048] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 10/25/2019] [Accepted: 10/28/2019] [Indexed: 12/16/2022]
Abstract
Microglia activation plays a key role in regulating inflammatory and immune reaction during cerebral ischemia and it exerts pro-inflammatory or anti-inflammatory effect depending on M1/M2 polarization phenotype. Cysteinyl leukotriene 2 receptor (CysLT2R) is a potent inflammatory mediator receptor, and involved in cerebral ischemic injury, but the mechanism of CysLT2R regulating inflammation and neuron damage remains unclear. Here, we found that LPS and CysLT2R agonist NMLTC4 significantly increased microglia proliferation and phagocytosis, up-regulated the mRNA expression of M1 polarization markers (IL-1β, TNF-α, IFN-γ, CD86 and iNOS), down-regulated the expression of M2 polarization markers (Arg-1, CD206, TGF-β, IL-10, Ym-1) and increased the release of IL-1β and TNF-α. CysLT2R selective antagonist HAMI3379 could antagonize these effects. IL-4 significantly up-regulated the mRNA expression of M2 polarization markers, and HAMI3379 further increased IL-4-induced up-regulation of M2 polarization markers expression. Additionally, LPS and NMLTC4 stimulated NF-κB p50 and p65 proteins expression, and promoted p50 transfer to the nucleus. Pre-treatment with HAMI3379 and NF-κB signaling inhibitor Bay 11-7082 could reverse the up-regulation of p50 and p65 proteins expression, and inhibited p50 transfer to the nucleus. The conditional medium of BV-2 cells contained HAMI3379 could inhibit SH-SY5Y cells apoptosis induced by LPS and NMLTC4. These results were further confirmed in primary microglia. The findings indicate that CysLT2R was involved in inflammation and neuronal damage by inducing the activation of microglia M1 polarization and NF-κB pathway, inhibiting microglia M1 polarization and promoting microglia polarization toward M2 phenotype which may exerts neuroprotective effects, and targeting CysLT2R may be a new therapeutic strategy against cerebral ischemia stroke.
Collapse
Affiliation(s)
- Rui Zhao
- Department of Pharmacy, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, No.3 Qingchun East Road, Hangzhou 310016, China
| | - Miaofa Ying
- Department of Pharmacy, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, No.3 Qingchun East Road, Hangzhou 310016, China
| | - Shenglong Gu
- Department of Pharmacy, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, No.3 Qingchun East Road, Hangzhou 310016, China
| | - Wei Yin
- Core Facilities, School of Medicine, Zhejiang University, 866 Yuhang Tang Road, Hangzhou 310058, China
| | - Yanwei Li
- Core Facilities, School of Medicine, Zhejiang University, 866 Yuhang Tang Road, Hangzhou 310058, China
| | - Hong Yuan
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhang Tang Road, Hangzhou 310058, China.
| | - Sanhua Fang
- Core Facilities, School of Medicine, Zhejiang University, 866 Yuhang Tang Road, Hangzhou 310058, China.
| | - Mingxing Li
- Department of Pharmacy, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, No.3 Qingchun East Road, Hangzhou 310016, China.
| |
Collapse
|
12
|
Biringer RG. The Role of Eicosanoids in Alzheimer's Disease. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2019; 16:ijerph16142560. [PMID: 31323750 PMCID: PMC6678666 DOI: 10.3390/ijerph16142560] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 07/09/2019] [Accepted: 07/13/2019] [Indexed: 12/21/2022]
Abstract
Alzheimer's disease (AD) is one of the most common neurodegenerative disorders known. Estimates from the Alzheimer's Association suggest that there are currently 5.8 million Americans living with the disease and that this will rise to 14 million by 2050. Research over the decades has revealed that AD pathology is complex and involves a number of cellular processes. In addition to the well-studied amyloid-β and tau pathology, oxidative damage to lipids and inflammation are also intimately involved. One aspect all these processes share is eicosanoid signaling. Eicosanoids are derived from polyunsaturated fatty acids by enzymatic or non-enzymatic means and serve as short-lived autocrine or paracrine agents. Some of these eicosanoids serve to exacerbate AD pathology while others serve to remediate AD pathology. A thorough understanding of eicosanoid signaling is paramount for understanding the underlying mechanisms and developing potential treatments for AD. In this review, eicosanoid metabolism is examined in terms of in vivo production, sites of production, receptor signaling, non-AD biological functions, and known participation in AD pathology.
Collapse
Affiliation(s)
- Roger G Biringer
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, 5000 Lakewood Ranch Blvd., Bradenton, FL 34211, USA.
| |
Collapse
|
13
|
Michael J, Marschallinger J, Aigner L. The leukotriene signaling pathway: a druggable target in Alzheimer’s disease. Drug Discov Today 2019; 24:505-516. [DOI: 10.1016/j.drudis.2018.09.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 08/17/2018] [Accepted: 09/12/2018] [Indexed: 12/29/2022]
|
14
|
Rahman SO, Singh RK, Hussain S, Akhtar M, Najmi AK. A novel therapeutic potential of cysteinyl leukotrienes and their receptors modulation in the neurological complications associated with Alzheimer's disease. Eur J Pharmacol 2018; 842:208-220. [PMID: 30389631 DOI: 10.1016/j.ejphar.2018.10.040] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 10/16/2018] [Accepted: 10/29/2018] [Indexed: 01/28/2023]
Abstract
Cysteinyl leukotrienes (cysLTs) are member of eicosanoid inflammatory lipid mediators family produced by oxidation of arachidonic acid by action of the enzyme 5-lipoxygenase (5-LOX). 5-LOX is activated by enzyme 5-Lipoxygenase-activating protein (FLAP), which further lead to production of cysLTs i.e. leukotriene C4 (LTC4), leukotriene D4 (LTD4) and leukotriene E4 (LTE4). CysLTs then produce their potent inflammatory actions by activating CysLT1 and CysLT2 receptors. Inhibitors of cysLTs are indicated in asthma, allergic rhinitis and other inflammatory disorders. Earlier studies have associated cysLTs and their receptors in several neurodegenerative disorders diseases like, multiple sclerosis, Parkinson's disease, Huntington's disease, epilepsy and Alzheimer's disease (AD). These inflammatory lipid mediators have previously shown effects on various aggravating factors of AD. However, not much data has been elucidated to test their role against AD clinically. Herein, through this review, we have provided the current and emerging information on the role of cysLTs and their receptors in various neurological complications responsible for the development of AD. In addition, literature evidences for the effect of cysLT inhibitors on distinct aspects of abnormalities in AD has also been reviewed. Promising advancement in understanding on the role of cysLTs on the various neuromodulatory processes and mechanisms may contribute to the development of newer and safer therapy for the treatment of AD in future.
Collapse
Affiliation(s)
- Syed Obaidur Rahman
- Pharmaceutical Medicine, Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India.
| | - Rakesh Kumar Singh
- School of Pharmaceutical Sciences, Apeejay Stya University, Sohna-Palwal Road, Sohna, Gurgaon 122013, Haryana, India.
| | - Salman Hussain
- Pharmaceutical Medicine, Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Mohd Akhtar
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India.
| | - Abul Kalam Najmi
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India.
| |
Collapse
|
15
|
Zhao B, Wang H, Li CX, Song SW, Fang SH, Wei EQ, Shi QJ. GPR17 mediates ischemia-like neuronal injury via microglial activation. Int J Mol Med 2018; 42:2750-2762. [PMID: 30226562 PMCID: PMC6192776 DOI: 10.3892/ijmm.2018.3848] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2018] [Accepted: 08/23/2018] [Indexed: 01/18/2023] Open
Abstract
GPR17 is a G (i)-coupled dual receptor, linked to P2Y and CysLT receptors stimulated by uracil nucleotides and cysteinyl leukotrienes, respectively. Recent evidence has demonstrated that GPR17 inhibition ameliorates the progression of cerebral ischemic injury by regulating neuronal death and microglial activation. The present study aimed to assess the detailed regulatory roles of this receptor in oxygen-glucose deprivation/recovery (OGD/R)-induced ischemia-like injury in vitro and explore the underlying mechanism. The results demonstrated that OGD/R induced ischemic neuronal injury and microglial activation, including enhanced phagocytosis and increased inflammatory cytokine release in neuron‑glial mixed cultures of cortical cells. GPR17 upregulation during OGD/R was spatially and temporally correlated with neuronal injury and microglial activation. In addition, GPR17 knockdown inhibited OGD/R-induced responses in neuron-glial mixed cultures. GPR17 knockdown also attenuated cell injury induced by the agonist leukotriene D4 (LTD4) or uridine 5′-diphosphate (UDP) in neuron-glial mixed cultures. However, GPR17 knockdown did not affect OGD/R-induced ischemic neuronal injury in primary cultures of neurons. In primary astrocyte cultures, neither GPR17 nor OGD/R induced injury. By contrast, GPR17 knockdown ameliorated OGD/R-induced microglial activation, boosting phagocytosis and inflammatory cytokine release in primary microglia cultures. Finally, the results demonstrated that the conditioned medium of microglia pretreated with OGD/R induced neuronal death, and the neuronal injury was significantly inhibited by GPR17 knockdown. These findings suggested that GPR17 may mediate ischemia-like neuronal injury and microglial activation in vitro; however, the protective effects on ischemic neuronal injury might depend upon microglial activation. Whether GPR17 regulates neuronal injury mediated by oligodendrocyte linkage remains to be investigated.
Collapse
Affiliation(s)
- Bing Zhao
- Department of Anesthesiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| | - Hao Wang
- Department of Neurology, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang 310012, P.R. China
| | - Cai-Xia Li
- Department of Anesthesiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| | - Sheng-Wen Song
- Department of Anesthesiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| | - San-Hua Fang
- Department of Pharmacology, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, P.R. China
| | - Er-Qing Wei
- Department of Pharmacology, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, P.R. China
| | - Qiao-Juan Shi
- Experimental Animal Center, Zhejiang Academy of Medical Sciences, Hangzhou, Zhejiang 310013, P.R. China
| |
Collapse
|
16
|
Freitas HR, Isaac AR, Malcher-Lopes R, Diaz BL, Trevenzoli IH, De Melo Reis RA. Polyunsaturated fatty acids and endocannabinoids in health and disease. Nutr Neurosci 2017; 21:695-714. [PMID: 28686542 DOI: 10.1080/1028415x.2017.1347373] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Polyunsaturated fatty acids (PUFAs) are lipid derivatives of omega-3 (docosahexaenoic acid, DHA, and eicosapentaenoic acid, EPA) or of omega-6 (arachidonic acid, ARA) synthesized from membrane phospholipids and used as a precursor for endocannabinoids (ECs). They mediate significant effects in the fine-tune adjustment of body homeostasis. Phyto- and synthetic cannabinoids also rule the daily life of billions worldwide, as they are involved in obesity, depression and drug addiction. Consequently, there is growing interest to reveal novel active compounds in this field. Cloning of cannabinoid receptors in the 90s and the identification of the endogenous mediators arachidonylethanolamide (anandamide, AEA) and 2-arachidonyglycerol (2-AG), led to the characterization of the endocannabinoid system (ECS), together with their metabolizing enzymes and membrane transporters. Today, the ECS is known to be involved in diverse functions such as appetite control, food intake, energy balance, neuroprotection, neurodegenerative diseases, stroke, mood disorders, emesis, modulation of pain, inflammatory responses, as well as in cancer therapy. Western diet as well as restriction of micronutrients and fatty acids, such as DHA, could be related to altered production of pro-inflammatory mediators (e.g. eicosanoids) and ECs, contributing to the progression of cardiovascular diseases, diabetes, obesity, depression or impairing conditions, such as Alzheimer' s disease. Here we review how diets based in PUFAs might be linked to ECS and to the maintenance of central and peripheral metabolism, brain plasticity, memory and learning, blood flow, and genesis of neural cells.
Collapse
Affiliation(s)
- Hércules Rezende Freitas
- a Laboratory of Neurochemistry, Institute of Biophysics Carlos Chagas Filho , Universidade Federal do Rio de Janeiro, Cidade Universitária , Ilha do Fundão, Rio de Janeiro , RJ 21941-902 , Brazil
| | - Alinny Rosendo Isaac
- a Laboratory of Neurochemistry, Institute of Biophysics Carlos Chagas Filho , Universidade Federal do Rio de Janeiro, Cidade Universitária , Ilha do Fundão, Rio de Janeiro , RJ 21941-902 , Brazil
| | | | - Bruno Lourenço Diaz
- c Laboratory of Inflammation, Institute of Biophysics Carlos Chagas Filho , Universidade Federal do Rio de Janeiro, Cidade Universitária , Ilha do Fundão, Rio de Janeiro , RJ 21941-902 , Brazil
| | - Isis Hara Trevenzoli
- d Laboratory of Molecular Endocrinology, Institute of Biophysics Carlos Chagas Filho , Universidade Federal do Rio de Janeiro, Cidade Universitária , Ilha do Fundão, Rio de Janeiro , RJ 21941-902 , Brazil
| | - Ricardo Augusto De Melo Reis
- a Laboratory of Neurochemistry, Institute of Biophysics Carlos Chagas Filho , Universidade Federal do Rio de Janeiro, Cidade Universitária , Ilha do Fundão, Rio de Janeiro , RJ 21941-902 , Brazil
| |
Collapse
|
17
|
Cysteinyl Leukotrienes as Potential Pharmacological Targets for Cerebral Diseases. Mediators Inflamm 2017; 2017:3454212. [PMID: 28607533 PMCID: PMC5451784 DOI: 10.1155/2017/3454212] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 04/10/2017] [Accepted: 04/19/2017] [Indexed: 02/06/2023] Open
Abstract
Cysteinyl leukotrienes (CysLTs) are potent lipid mediators widely known for their actions in asthma and in allergic rhinitis. Accumulating data highlights their involvement in a broader range of inflammation-associated diseases such as cancer, atopic dermatitis, rheumatoid arthritis, and cardiovascular diseases. The reported elevated levels of CysLTs in acute and chronic brain lesions, the association between the genetic polymorphisms in the LTs biosynthesis pathways and the risk of cerebral pathological events, and the evidence from animal models link also CysLTs and brain diseases. This review will give an overview of how far research has gone into the evaluation of the role of CysLTs in the most prevalent neurodegenerative disorders (ischemia, Alzheimer's and Parkinson's diseases, multiple sclerosis/experimental autoimmune encephalomyelitis, and epilepsy) in order to understand the underlying mechanism by which they might be central in the disease progression.
Collapse
|
18
|
Hoxha M, Rovati GE, Cavanillas AB. The leukotriene receptor antagonist montelukast and its possible role in the cardiovascular field. Eur J Clin Pharmacol 2017; 73:799-809. [PMID: 28374082 DOI: 10.1007/s00228-017-2242-2] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 03/22/2017] [Indexed: 12/31/2022]
Abstract
BACKGROUND Cysteinyl leukotrienes (LTC4, LTD4, and LTE4) are pro-inflammatory mediators of the 5-lipooxygenase (5-LO) pathway, that play an important role in bronchoconstriction, but can also enhance endothelial cell permeability and myocardial contractility, and are involved in many other inflammatory conditions. In the late 1990s, leukotriene receptor antagonists (LTRAs) were introduced in therapy for asthma and later on, approved for the relief of the symptoms of allergic rhinitis, chronic obstructive pulmonary disease, and urticaria. In addition, it has been shown that LTRAs may have a potential role in preventing atherosclerosis progression. PURPOSE The aims of this short review are to delineate the potential cardiovascular protective role of a LTRA, montelukast, beyond its traditional use, and to foster the design of appropriate clinical trials to test this hypothesis. RESULTS AND CONCLUSIONS What it is known about leukotriene receptor antagonists? •Leukotriene receptor antagonist, such as montelukast and zafirlukast, is used in asthma, COPD, and allergic rhinitis. • Montelukast is the most prescribed CysLT1 antagonist used in asthmatic patients. • Different in vivo animal studies have shown that leukotriene receptor antagonists can prevent the atherosclerosis progression, and have a protective role after cerebral ischemia. What we still need to know? • Today, there is a need for conducting clinical trials to assess the role of montelukast in reducing cardiovascular risk and to further understand the mechanism of action behind this effect.
Collapse
Affiliation(s)
- Malvina Hoxha
- Department of Chemical, Toxicological and Pharmacological Evaluation of Drugs, Catholic University Our Lady of Good Counsel, Rruga. D. Hoxha, Tirana, Albania.
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Via Balzaretti, 9-20133, Milan, Italy.
| | - G Enrico Rovati
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Via Balzaretti, 9-20133, Milan, Italy
| | - Aurora Bueno Cavanillas
- IBS Granada, University of Granada, CIBER of Epidemiology and Public Health (CIBERESP), Granada, Spain
| |
Collapse
|
19
|
Ghosh A, Chen F, Thakur A, Hong H. Cysteinyl Leukotrienes and Their Receptors: Emerging Therapeutic Targets in Central Nervous System Disorders. CNS Neurosci Ther 2016; 22:943-951. [PMID: 27542570 DOI: 10.1111/cns.12596] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Revised: 07/14/2016] [Accepted: 07/18/2016] [Indexed: 12/17/2022] Open
Abstract
Cysteinyl leukotrienes are a group of the inflammatory lipid molecules well known as mediators of inflammatory signaling in the allergic diseases. Although they are traditionally known for their role in allergic asthma, allergic rhinitis, and others, recent advances in the field of biomedical research highlighted the role of these inflammatory mediators in a broader range of diseases such as in the inflammation associated with the central nervous system (CNS) disorders, vascular inflammation (atherosclerotic), and in cancer. Among the CNS diseases, they, along with their synthesis precursor enzyme 5-lipoxygenase and their receptors, have been shown to be associated with brain injury, Multiple sclerosis, Alzheimer's disease, Parkinson's disease, brain ischemia, epilepsy, and others. However, a lot more remains elusive as the research in these areas is emerging and only a little has been discovered. Herein, through this review, we first provided a general up-to-date information on the synthesis pathway and the receptors for the molecules. Next, we summarized the current findings on their role in the brain disorders, with an insight given to the future perspectives.
Collapse
Affiliation(s)
- Arijit Ghosh
- Laboratory for Alzheimer's Disease and Related Disorders, Department of Pharmacology, China Pharmaceutical University, Nanjing, China.,Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing, China
| | - Fang Chen
- Laboratory for Alzheimer's Disease and Related Disorders, Department of Pharmacology, China Pharmaceutical University, Nanjing, China.,Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing, China
| | - Abhimanyu Thakur
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong SAR, China
| | - Hao Hong
- Laboratory for Alzheimer's Disease and Related Disorders, Department of Pharmacology, China Pharmaceutical University, Nanjing, China.,Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
20
|
Ma Y, Wang J, Wang Y, Yang GY. The biphasic function of microglia in ischemic stroke. Prog Neurobiol 2016; 157:247-272. [PMID: 26851161 DOI: 10.1016/j.pneurobio.2016.01.005] [Citation(s) in RCA: 519] [Impact Index Per Article: 57.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 12/22/2015] [Accepted: 01/10/2016] [Indexed: 12/16/2022]
Abstract
Microglia are brain resident macrophages originated from primitive progenitor cells in the yolk sac. Microglia can be activated within hours and recruited to the lesion site. Traditionally, microglia activation is considered to play a deleterious role in ischemic stroke, as inhibition of microglia activation attenuates ischemia induced brain injury. However, increasing evidence show that microglia activation is critical for attenuating neuronal apoptosis, enhancing neurogenesis, and promoting functional recovery after cerebral ischemia. Differential polarization of microglia could likely explain the biphasic role of microglia in ischemia. We comprehensively reviewed the mechanisms involved in regulating microglia activation and polarization. The latest discoveries of microRNAs in modulating microglia function are discussed. In addition, the interaction between microglia and other cells including neurons, astrocytes, oligodendrocytes, and stem cells were also reviewed. Future therapies targeting microglia may not exclusively aim at suppressing microglia activation, but also at modulating microglia polarization at different stages of ischemic stroke. More work is needed to elucidate the cellular and molecular mechanisms of microglia polarization under ischemic environment. The roles of microRNAs and transplanted stem cells in mediating microglia activation and polarization during brain ischemia also need to be further studied.
Collapse
Affiliation(s)
- Yuanyuan Ma
- Department of Neurology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China; Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Jixian Wang
- Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China; Department of Rehabilitation, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China
| | - Yongting Wang
- Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China.
| | - Guo-Yuan Yang
- Department of Neurology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China; Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China.
| |
Collapse
|
21
|
CysLT 2 receptor mediates lipopolysaccharide-induced microglial inflammation and consequent neurotoxicity in vitro. Brain Res 2015; 1624:433-445. [DOI: 10.1016/j.brainres.2015.08.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Revised: 07/17/2015] [Accepted: 08/07/2015] [Indexed: 01/01/2023]
|
22
|
Vella J, Zammit C, Di Giovanni G, Muscat R, Valentino M. The central role of aquaporins in the pathophysiology of ischemic stroke. Front Cell Neurosci 2015; 9:108. [PMID: 25904843 PMCID: PMC4389728 DOI: 10.3389/fncel.2015.00108] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 04/10/2015] [Indexed: 11/16/2022] Open
Abstract
Stroke is a complex and devastating neurological condition with limited treatment options. Brain edema is a serious complication of stroke. Early edema formation can significantly contribute to infarct formation and thus represents a promising target. Aquaporin (AQP) water channels contribute to water homeostasis by regulating water transport and are implicated in several disease pathways. At least 7 AQP subtypes have been identified in the rodent brain and the use of transgenic mice has greatly aided our understanding of their functions. AQP4, the most abundant channel in the brain, is up-regulated around the peri-infarct border in transient cerebral ischemia and AQP4 knockout mice demonstrate significantly reduced cerebral edema and improved neurological outcome. In models of vasogenic edema, brain swelling is more pronounced in AQP4-null mice than wild-type providing strong evidence of the dual role of AQP4 in the formation and resolution of both vasogenic and cytotoxic edema. AQP4 is co-localized with inwardly rectifying K(+)-channels (Kir4.1) and glial K(+) uptake is attenuated in AQP4 knockout mice compared to wild-type, indicating some form of functional interaction. AQP4-null mice also exhibit a reduction in calcium signaling, suggesting that this channel may also be involved in triggering pathological downstream signaling events. Associations with the gap junction protein Cx43 possibly recapitulate its role in edema dissipation within the astroglial syncytium. Other roles ascribed to AQP4 include facilitation of astrocyte migration, glial scar formation, modulation of inflammation and signaling functions. Treatment of ischemic cerebral edema is based on the various mechanisms in which fluid content in different brain compartments can be modified. The identification of modulators and inhibitors of AQP4 offer new therapeutic avenues in the hope of reducing the extent of morbidity and mortality in stroke.
Collapse
Affiliation(s)
| | | | | | | | - Mario Valentino
- Department of Physiology and Biochemistry, University of MaltaMsida, Malta
| |
Collapse
|
23
|
Shi QJ, Wang H, Liu ZX, Fang SH, Song XM, Lu YB, Zhang WP, Sa XY, Ying HZ, Wei EQ. HAMI 3379, a CysLT2R antagonist, dose- and time-dependently attenuates brain injury and inhibits microglial inflammation after focal cerebral ischemia in rats. Neuroscience 2015; 291:53-69. [PMID: 25681271 DOI: 10.1016/j.neuroscience.2015.02.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Revised: 01/19/2015] [Accepted: 02/02/2015] [Indexed: 12/29/2022]
Abstract
Cysteinyl leukotrienes (CysLTs) induce inflammatory responses by activating their receptors, CysLT1R and CysLT2R. We have reported that CysLT2R is involved in neuronal injury, astrocytosis, and microgliosis, and that intracerebroventricular (i.c.v.) injection of the selective CysLT2R antagonist HAMI 3379 protects against acute brain injury after focal cerebral ischemia in rats. In the present study, we clarified features of the protective effect of intraperitoneally-injected HAMI 3379 in rats. We found that HAMI 3379 attenuated the acute brain injury 24 h after middle cerebral artery occlusion (MCAO) with effective doses of 0.1-0.4 mg/kg and a therapeutic window of ∼1h. It attenuated the neurological deficits, and reduced infarct volume, brain edema, and neuronal loss and degeneration 24 and 72h after MCAO. RNA interference with i.c.v. injection of CysLT2R short hairpin RNA (shRNA) attenuated the acute injury as well. Also, HAMI 3379 inhibited release of the cytokines IL-1β, interferon-γ (IFN-γ), and tumor necrosis factor-α (TNF-α) into the serum and cerebrospinal fluid 24h after MCAO. Moreover, HAMI 3379 ameliorated the microglial activation and neutrophil accumulation in the ischemic regions, but did not affect astrocyte proliferation 72h after MCAO. In comparison, the CysLT1R antagonist pranlukast did not affect microglial activation and IFN-γ release, but inhibited astrocyte proliferation and reduced serum IL-4. Thus, we conclude that HAMI 3379 has a protective effect on acute and subacute ischemic brain injury, and attenuates microglia-related inflammation. CysLT2R antagonist(s) alone or in combination with CysLT1R antagonists may be a novel class of therapeutic agents in the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Q J Shi
- Department of Pharmacology, School of Medicine, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China; Experimental Animal Center, Zhejiang Academy of Medical Sciences, 182 Tianmushan Road, Hangzhou 310013, China
| | - H Wang
- Department of Neurology, Tongde Hospital of Zhejiang Province, Hangzhou 310012, China
| | - Z X Liu
- Department of Pharmacology, School of Medicine, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
| | - S H Fang
- Department of Pharmacology, School of Medicine, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
| | - X M Song
- Experimental Animal Center, Zhejiang Academy of Medical Sciences, 182 Tianmushan Road, Hangzhou 310013, China
| | - Y B Lu
- Department of Pharmacology, School of Medicine, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
| | - W P Zhang
- Department of Pharmacology, School of Medicine, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
| | - X Y Sa
- Experimental Animal Center, Zhejiang Academy of Medical Sciences, 182 Tianmushan Road, Hangzhou 310013, China
| | - H Z Ying
- Experimental Animal Center, Zhejiang Academy of Medical Sciences, 182 Tianmushan Road, Hangzhou 310013, China
| | - E Q Wei
- Department of Pharmacology, School of Medicine, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China.
| |
Collapse
|
24
|
Bäck M, Powell WS, Dahlén SE, Drazen JM, Evans JF, Serhan CN, Shimizu T, Yokomizo T, Rovati GE. Update on leukotriene, lipoxin and oxoeicosanoid receptors: IUPHAR Review 7. Br J Pharmacol 2014; 171:3551-74. [PMID: 24588652 DOI: 10.1111/bph.12665] [Citation(s) in RCA: 155] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Revised: 02/06/2014] [Accepted: 02/18/2014] [Indexed: 12/14/2022] Open
Abstract
The endogenous ligands for the LT, lipoxin (LX) and oxoeicosanoid receptors are bioactive products produced by the action of the lipoxygenase family of enzymes. The LT receptors BLT1 and BLT2 , are activated by LTB4 and the CysLT1 and CysLT2 receptors are activated by the cysteinyl-LTs, whereas oxoeicosanoids exert their action through the OXE receptor. In contrast to these pro-inflammatory mediators, LXA4 transduces responses associated with the resolution of inflammation through the receptor FPR2/ALX (ALX/FPR2). The aim of the present review is to give a state of the field on these receptors, with focus on recent important findings. For example, BLT1 receptor signalling in cancer and the dual role of the BLT2 receptor in pro- and anti-inflammatory actions have added more complexity to lipid mediator signalling. Furthermore, a cross-talk between the CysLT and P2Y receptor systems has been described, and also the presence of novel receptors for cysteinyl-LTs, such as GPR17 and GPR99. Finally, lipoxygenase metabolites derived from ω-3 essential polyunsaturated acids, the resolvins, activate the receptors GPR32 and ChemR23. In conclusion, the receptors for the lipoxygenase products make up a sophisticated and tightly controlled system of endogenous pro- and anti-inflammatory signalling in physiology and pathology.
Collapse
Affiliation(s)
- Magnus Bäck
- Nomenclature Subcommittee for Leukotriene Receptors, International Union of Basic and Clinical Pharmacology, Stockholm, Sweden; Department of Medicine, Karolinska Institutet, Stockholm, Sweden; Department of Cardiology, Karolinska University Hospital, Stockholm, Sweden
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Bertin J, Jalaguier P, Barat C, Roy MA, Tremblay MJ. Exposure of human astrocytes to leukotriene C4 promotes a CX3CL1/fractalkine-mediated transmigration of HIV-1-infected CD4⁺ T cells across an in vitro blood-brain barrier model. Virology 2014; 454-455:128-38. [PMID: 24725939 DOI: 10.1016/j.virol.2014.02.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Revised: 12/23/2013] [Accepted: 02/07/2014] [Indexed: 11/18/2022]
Abstract
Eicosanoids, including cysteinylleukotrienes (cysLTs), are found in the central nervous system (CNS) of individuals infected with HIV-1. Few studies have addressed the contribution of cysLTs in HIV-1-associated CNS disorders. We demonstrate that conditioned medium from human astrocytes treated with leukotriene C4 (LTC4) increases the transmigration of HIV-1-infected CD4(+) T cells across an in vitro blood-brain barrier (BBB) model using cultured brain endothelial cells. Additional studies indicate that the higher cell migration is linked with secretion by astrocytes of CX3CL1/fractalkine, a chemokine that has chemoattractant activity for CD4(+) T cells. Moreover, we report that the enhanced cell migration across BBB leads to a more important CD4(+) T cell-mediated HIV-1 transfer toward macrophages. Altogether data presented in the present study reveal the important role that LTC4, a metabolite of arachidonic acid, may play in the HIV-1-induced neuroinvasion, neuropathogenesis and disease progression.
Collapse
Affiliation(s)
- Jonathan Bertin
- Axe des Maladies Infectieuses et Immunitaires, Centre de recherche du Centre Hospitalier Universitaire (CHU) de Québec - pavillon CHUL, Canada
| | - Pascal Jalaguier
- Axe des Maladies Infectieuses et Immunitaires, Centre de recherche du Centre Hospitalier Universitaire (CHU) de Québec - pavillon CHUL, Canada
| | - Corinne Barat
- Axe des Maladies Infectieuses et Immunitaires, Centre de recherche du Centre Hospitalier Universitaire (CHU) de Québec - pavillon CHUL, Canada
| | - Marc-André Roy
- Axe des Maladies Infectieuses et Immunitaires, Centre de recherche du Centre Hospitalier Universitaire (CHU) de Québec - pavillon CHUL, Canada
| | - Michel J Tremblay
- Axe des Maladies Infectieuses et Immunitaires, Centre de recherche du Centre Hospitalier Universitaire (CHU) de Québec - pavillon CHUL, Canada; Département de Microbiologie-Infectiologie et Immunologie, Faculté de médecine, Université Laval, Québec, Canada.
| |
Collapse
|
26
|
Cysteinyl leukotriene receptor 1 mediates LTD4-induced activation of mouse microglial cells in vitro. Acta Pharmacol Sin 2014; 35:33-40. [PMID: 24141567 DOI: 10.1038/aps.2013.130] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Accepted: 08/22/2013] [Indexed: 01/03/2023] Open
Abstract
AIM To investigate the roles of cysteinyl leukotriene receptors CysLT1R and CysLT2R in leukotriene D4 (LTD4)-induced activation of microglial cells in vitro. METHODS Mouse microglial cell line BV2 was transfected with pcDNA3.1(+)-hCysLT1R or pcDNA3.1(+)-hCysLT2R. The expression of relevant mRNAs and proteins in the cells was detected using RT-PCR and Western blotting, respectively. Phagocytosis was determined with flow cytometry analysis. The release of interleukin-1β (IL-1β) from the cells was measured using an ELISA assay. RESULTS The expression of CysLT1R or CysLT2R was considerably increased in the transfected BV2 cells, and the receptors were mainly distributed in the plasma membrane and cytosol. Treatment of the cells expressing CysLT1R or CysLT2R with CysLT receptor agonist LTD4 (0.1-100 nmol/L) concentration-dependently enhanced the phagocytosis, and increased mRNA expression and release of IL-1β. Moreover, the responses of hCysLT1R-BV2 cells to LTD4 were significantly larger than those of hCysLT2R-BV2 or WT-BV2 cells. Pretreatment of hCysLT1R-BV2 cells with the selective CysLT1R antagonist montelukast (1 μmol/L) significantly blocked LTD4-induced phagocytosis as well as the mRNA expression and release of IL-1β, whereas the selective CysLT2R antagonist HAMI 3379 (1 μmol/L) had no such effects. CONCLUSION CysLT1R mediates LTD4-induced activation of BV2 cells, suggesting that CysLT1R antagonists may exert anti-inflammatory activity in brain diseases.
Collapse
|
27
|
Zhao B, Zhang M, Han X, Zhang XY, Xing Q, Dong X, Shi QJ, Huang P, Lu YB, Wei EQ, Xia Q, Zhang WP, Tang C. Cerebral ischemia is exacerbated by extracellular nicotinamide phosphoribosyltransferase via a non-enzymatic mechanism. PLoS One 2013; 8:e85403. [PMID: 24392007 PMCID: PMC3877362 DOI: 10.1371/journal.pone.0085403] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Accepted: 11/26/2013] [Indexed: 11/18/2022] Open
Abstract
Intracellular nicotinamide phosphoribosyltransferase (iNAMPT) in neuron has been known as a protective factor against cerebral ischemia through its enzymatic activity, but the role of central extracellular NAMPT (eNAMPT) is not clear. Here we show that eNAMPT protein level was elevated in the ischemic rat brain after middle-cerebral-artery occlusion (MCAO) and reperfusion, which can be traced to at least in part from blood circulation. Administration of recombinant NAMPT protein exacerbated MCAO-induced neuronal injury in rat brain, while exacerbated oxygen-glucose-deprivation (OGD) induced neuronal injury only in neuron-glial mixed culture, but not in neuron culture. In the mixed culture, NAMPT protein promoted TNF-α release in a time- and concentration-dependent fashion, while TNF-α neutralizing antibody protected OGD-induced, NAMPT-enhanced neuronal injury. Importantly, H247A mutant of NAMPT with essentially no enzymatic activity exerted similar effects on ischemic neuronal injury and TNF-α release as the wild type protein. Thus, eNAMPT is an injurious and inflammatory factor in cerebral ischemia and aggravates ischemic neuronal injury by triggering TNF-α release from glia cells, via a mechanism not related to NAMPT enzymatic activity.
Collapse
Affiliation(s)
- Bing Zhao
- Department of Pharmacology, Key Laboratory of Medical Neurobiology of Ministry of Health of China, Zhejiang Province Key Laboratory of Neurobiology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Anesthesiology, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Meng Zhang
- Department of Physiology, Zhejiang University School of Medicine, Hangzhou, China
| | - Xue Han
- Department of Pharmacology, Key Laboratory of Medical Neurobiology of Ministry of Health of China, Zhejiang Province Key Laboratory of Neurobiology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xia-Yan Zhang
- Department of Pharmacology, Key Laboratory of Medical Neurobiology of Ministry of Health of China, Zhejiang Province Key Laboratory of Neurobiology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Qiong Xing
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Wuhan Center for Magnetic, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, China
| | - Xu Dong
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Wuhan Center for Magnetic, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, China
| | - Qiao-Juan Shi
- Department of Pharmacology, Key Laboratory of Medical Neurobiology of Ministry of Health of China, Zhejiang Province Key Laboratory of Neurobiology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Peng Huang
- Department of Pharmacology, Key Laboratory of Medical Neurobiology of Ministry of Health of China, Zhejiang Province Key Laboratory of Neurobiology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yun-Bi Lu
- Department of Pharmacology, Key Laboratory of Medical Neurobiology of Ministry of Health of China, Zhejiang Province Key Laboratory of Neurobiology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Er-Qing Wei
- Department of Pharmacology, Key Laboratory of Medical Neurobiology of Ministry of Health of China, Zhejiang Province Key Laboratory of Neurobiology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Qiang Xia
- Department of Physiology, Zhejiang University School of Medicine, Hangzhou, China
| | - Wei-Ping Zhang
- Department of Pharmacology, Key Laboratory of Medical Neurobiology of Ministry of Health of China, Zhejiang Province Key Laboratory of Neurobiology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- * E-mail:
| | - Chun Tang
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Wuhan Center for Magnetic, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, China
| |
Collapse
|
28
|
Li Y, Liu Z, Xin H, Chopp M. The role of astrocytes in mediating exogenous cell-based restorative therapy for stroke. Glia 2013; 62:1-16. [PMID: 24272702 DOI: 10.1002/glia.22585] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Revised: 08/08/2013] [Accepted: 09/18/2013] [Indexed: 12/19/2022]
Abstract
Astrocytes have not been a major therapeutic target for the treatment of stroke, with most research emphasis on the neuron. Given the essential role that astrocytes play in maintaining physiological function of the central nervous system and the very rapid and sensitive reaction astrocytes have in response to cerebral injury or ischemic insult, we propose to replace the neurocentric view for treatment with a more nuanced astrocytic centered approach. In addition, after decades of effort in attempting to develop neuroprotective therapies, which target reduction of the ischemic lesion, there are no effective clinical treatments for stroke, aside from thrombolysis with tissue plasminogen activator, which is used in a small minority of patients. A more promising therapeutic approach, which may affect nearly all stroke patients, may be in promoting endogenous restorative mechanisms, which enhance neurological recovery. A focus of efforts in stimulating recovery post stroke is the use of exogenously administered cells. The present review focuses on the role of the astrocyte in mediating the brain network, brain plasticity, and neurological recovery post stroke. As a model to describe the interaction of a restorative cell-based therapy with astrocytes, which drives recovery from stroke, we specifically highlight the subacute treatment of stroke with multipotent mesenchymal stromal cell therapy.
Collapse
Affiliation(s)
- Yi Li
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan
| | | | | | | |
Collapse
|
29
|
Yagami T, Yamamoto Y, Kohma H. Leukotriene receptor antagonists, LY293111 and ONO-1078, protect neurons from the sPLA2-IB-induced neuronal cell death independently of blocking their receptors. Neurochem Int 2013; 63:163-71. [DOI: 10.1016/j.neuint.2013.06.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2013] [Revised: 06/06/2013] [Accepted: 06/11/2013] [Indexed: 12/29/2022]
|
30
|
Zhang XY, Wang XR, Xu DM, Yu SY, Shi QJ, Zhang LH, Chen L, Fang SH, Lu YB, Zhang WP, Wei EQ. HAMI 3379, a CysLT2 Receptor Antagonist, Attenuates Ischemia-Like Neuronal Injury by Inhibiting Microglial Activation. J Pharmacol Exp Ther 2013; 346:328-41. [DOI: 10.1124/jpet.113.203604] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
|
31
|
Ström JO, Strid T, Hammarström S. Disruption of the alox5ap gene ameliorates focal ischemic stroke: possible consequence of impaired leukotriene biosynthesis. BMC Neurosci 2012. [PMID: 23194405 PMCID: PMC3557197 DOI: 10.1186/1471-2202-13-146] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Background Leukotrienes are potent inflammatory mediators, which in a number of studies have been found to be associated with ischemic stroke pathology: gene variants affecting leukotriene synthesis, including the FLAP (ALOX5AP) gene, have in human studies shown correlation to stroke incidence, and animal studies have demonstrated protective properties of various leukotriene-disrupting drugs. However, no study has hitherto described a significant effect of a genetic manipulation of the leukotriene system on ischemic stroke. Therefore, we decided to compare the damage from focal cerebral ischemia between wild type and FLAP knockout mice. Damage was evaluated by infarct staining and a functional test after middle cerebral artery occlusion in 20 wild type and 20 knockout male mice. Results Mortality-adjusted median infarct size was 18.4 (3.2-76.7) mm3 in the knockout group, compared to 72.0 (16.7-174.0) mm3 in the wild type group (p < 0.0005). There was also a tendency of improved functional score in the knockout group (p = 0.068). Analysis of bone marrow cells confirmed that knockout animals had lost their ability to form leukotrienes. Conclusions Since the local inflammatory reaction after ischemic stroke is known to contribute to the brain tissue damage, the group difference seen in the current study could be a consequence of a milder inflammatory reaction in the knockout group. Our results add evidence to the notion that leukotrienes are important in ischemic stroke, and that blocked leukotriene production ameliorates cerebral damage.
Collapse
Affiliation(s)
- Jakob O Ström
- Division of Clinical Chemistry, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | | | | |
Collapse
|
32
|
Aggravated inflammation and increased expression of cysteinyl leukotriene receptors in the brain after focal cerebral ischemia in AQP4-deficient mice. Neurosci Bull 2012; 28:680-92. [PMID: 23132680 DOI: 10.1007/s12264-012-1281-z] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2012] [Accepted: 04/06/2012] [Indexed: 10/27/2022] Open
Abstract
OBJECTIVE Aquaporin-4 (AQP4), the main water channel protein in the brain, plays a critical role in water homeostasis and brain edema. Here, we investigated its role in the inflammatory responses after focal cerebral ischemia. METHODS In AQP4-knockout (KO) and wild-type mice, focal cerebral ischemia was induced by 30 min of middle cerebral arterial occlusion (MCAO). Ischemic neuronal injury and cellular inflammatory responses, as well as the expression and localization of cysteinyl leukotriene CysLT(2) and CysLT(1) receptors, were determined at 24 and 72 h after MCAO. RESULTS AQP4-KO mice showed more neuronal loss, more severe microglial activation and neutrophil infiltration, but less astrocyte proliferation in the brain after MCAO than wild-type mice. In addition, the protein levels of both CysLT(1) and CysLT(2) receptors were up-regulated in the ischemic brain, and the up-regulation was more pronounced in AQP4-KO mice. The CysLT(1) and CysLT(2) receptors were primarily localized in neurons, microglia and neutrophils; those localized in microglia and neutrophils were enhanced in AQP4-KO mice. CONCLUSION AQP4 may play an inhibitory role in postischemic inflammation.
Collapse
|
33
|
Shi QJ, Xiao L, Zhao B, Zhang XY, Wang XR, Xu DM, Yu SY, Fang SH, Lu YB, Zhang WP, Sa XY, Wei EQ. Intracerebroventricular injection of HAMI 3379, a selective cysteinyl leukotriene receptor 2 antagonist, protects against acute brain injury after focal cerebral ischemia in rats. Brain Res 2012; 1484:57-67. [PMID: 23000196 DOI: 10.1016/j.brainres.2012.09.020] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2012] [Revised: 09/08/2012] [Accepted: 09/10/2012] [Indexed: 01/28/2023]
Abstract
Cysteinyl leukotrienes (CysLTs) induce inflammatory responses by activating their receptors, CysLT(1)R and CysLT(2)R. We recently reported that CysLT(2)R is involved in neuronal injury, astrocytosis and microgliosis after focal cerebral ischemia in rats. Here, we determined whether HAMI 3379, a selective CysLT(2)R antagonist, protects against acute brain injury after focal cerebral ischemia in rats. We induced transient focal cerebral ischemia by 30 min of middle cerebral artery occlusion (MCAO), followed by 24h of reperfusion. HAMI 3379 (1, 10 or 100 ng) was injected intracerebroventricularly (i.c.v.) 30 min before MCAO, and the CysLT(1)R antagonist pranlukast (0.1mg/kg, i.p.) was used as a positive control. HAMI 3379 at 10 and 100 ng (but not at 1 ng) attenuated the neurological deficits, and reduced infarct volume, brain edema, IgG exudation, neuronal degeneration and neuronal loss. This protective effect was similar to that of pranlukast. Thus, HAMI 3339 at 10-100 ng i.c.v. is neuroprotective against acute brain injury after focal cerebral ischemia in rats. These findings suggest therapeutic potential for CysLT(2)R antagonists in the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Qiao-Juan Shi
- Department of Pharmacology, School of Medicine, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Huang XQ, Zhang XY, Wang XR, Yu SY, Fang SH, Lu YB, Zhang WP, Wei EQ. Transforming growth factor β1-induced astrocyte migration is mediated in part by activating 5-lipoxygenase and cysteinyl leukotriene receptor 1. J Neuroinflammation 2012; 9:145. [PMID: 22734808 PMCID: PMC3419068 DOI: 10.1186/1742-2094-9-145] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2012] [Accepted: 05/17/2012] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Transforming growth factor-β 1 (TGF-β 1) is an important regulator of cell migration and plays a role in the scarring response in injured brain. It is also reported that 5-lipoxygenase (5-LOX) and its products, cysteinyl leukotrienes (CysLTs, namely LTC₄, LTD₄ and LTE₄), as well as cysteinyl leukotriene receptor 1 (CysLT₁R) are closely associated with astrocyte proliferation and glial scar formation after brain injury. However, how these molecules act on astrocyte migration, an initial step of the scarring response, is unknown. To clarify this, we determined the roles of 5-LOX and CysLT₁R in TGF-β 1-induced astrocyte migration. METHODS In primary cultures of rat astrocytes, the effects of TGF-β 1 and CysLT receptor agonists on migration and proliferation were assayed, and the expression of 5-LOX, CysLT receptors and TGF-β1 was detected. 5-LOX activation was analyzed by measuring its products (CysLTs) and applying its inhibitor. The role of CysLT₁R was investigated by applying CysLT receptor antagonists and CysLT₁R knockdown by small interfering RNA (siRNA). TGF-β 1 release was assayed as well. RESULTS TGF-β 1-induced astrocyte migration was potentiated by LTD₄, but attenuated by the 5-LOX inhibitor zileuton and the CysLT₁R antagonist montelukast. The non-selective agonist LTD₄ at 0.1 to 10 nM also induced a mild migration; however, the selective agonist N-methyl-LTC₄ and the selective antagonist Bay cysLT2 for CysLT₂R had no effects. Moreover, CysLT₁R siRNA inhibited TGF-β 1- and LTD₄-induced astrocyte migration by down-regulating the expression of this receptor. However, TGF-β 1 and LTD4 at various concentrations did not affect astrocyte proliferation 24 h after exposure. On the other hand, TGF-β 1 increased 5-LOX expression and the production of CysLTs, and up-regulated CysLT1R (not CysLT₂R), while LTD4 and N-methyl-LTC4 did not affect TGF-β 1 expression and release. CONCLUSIONS TGF-β 1-induced astrocyte migration is, at least in part, mediated by enhanced endogenous CysLTs through activating CysLT₁R. These findings indicate that the interaction between the cytokine TGF-β 1 and the pro-inflammatory mediators CysLTs in the regulation of astrocyte function is relevant to glial scar formation.
Collapse
Affiliation(s)
- Xue-Qin Huang
- Department of Pharmacology, Key Laboratory of Medical Neurobiology of Ministry of Health of China, Zhejiang Province Key Laboratory of Neurobiology, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou 310058, China
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Zhao B, Zhao CZ, Zhang XY, Huang XQ, Shi WZ, Fang SH, Lu YB, Zhang WP, Xia Q, Wei EQ. The new P2Y-like receptor G protein-coupled receptor 17 mediates acute neuronal injury and late microgliosis after focal cerebral ischemia in rats. Neuroscience 2011; 202:42-57. [PMID: 22155652 DOI: 10.1016/j.neuroscience.2011.11.066] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2011] [Revised: 11/25/2011] [Accepted: 11/28/2011] [Indexed: 12/29/2022]
Abstract
G protein-coupled receptor 17 (GPR17), the new P2Y-like receptor, is phylogenetically related to the P2Y and cysteinyl leukotriene receptors, and responds to both uracil nucleotides and cysteinyl leukotrienes. GPR17 has been proposed to be a damage sensor in ischemic stroke; however, its role in brain inflammation needs further detailed investigation. Here, we extended previous studies on the spatiotemporal profiles of GPR17 expression and localization, and their implications for brain injury after focal cerebral ischemia. We found that in the ischemic core, GPR17 mRNA and protein levels were upregulated at both 12-24 h and 7-14 days, but in the boundary zone the levels increased 7-14 days after reperfusion. The spatiotemporal pattern of GPR17 expression well matched the acute and late (subacute/chronic) responses in the ischemic brain. According to previous findings, in the acute phase, after ischemia (24 h), upregulated GPR17 was localized in injured neurons in the ischemic core and in a few microglia in the ischemic core and boundary zone. In the late phase (14 days), it was localized in microglia, especially in activated (ED1-positive) microglia in the ischemic core, but weakly in most microglia in the boundary zone. No GPR17 was detectable in astrocytes. GPR17 knockdown by a small interfering RNA attenuated the neurological dysfunction, infarction, and neuron loss at 24 h, and brain atrophy, neuron loss, and microglial activation at 14 days after reperfusion. Thus, GPR17 might mediate acute neuronal injury and late microgliosis after focal cerebral ischemia.
Collapse
Affiliation(s)
- B Zhao
- Department of Pharmacology, School of Medicine, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|