Skip to main content
CNS Neuroscience & Therapeutics logoLink to CNS Neuroscience & Therapeutics
. 2016 Aug 19;22(12):943–951. doi: 10.1111/cns.12596

Cysteinyl Leukotrienes and Their Receptors: Emerging Therapeutic Targets in Central Nervous System Disorders

Arijit Ghosh 1,2, Fang Chen 1,2, Abhimanyu Thakur 3, Hao Hong 1,2,
PMCID: PMC6492851  PMID: 27542570

Summary

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.

Keywords: Alzheimer's disease, Cerebral ischemia, Cysteinyl leukotrienes type 1 receptor, G‐protein‐coupled receptor 17, Multiple sclerosis

Background

The importance of the lipid inflammatory mediators in health and disease has been emerging area of interest. Leukotrienes (LTs), along with prostaglandins, thromboxanes, and lipoxins, are the major constituents of a group of biologically active, oxygenated, polyunsaturated, long‐chain fatty acids, known as the eicosanoids. They are secreted by mast cells, eosinophil, and leukocytes during inflammation and possess a wide range of biological activities such as leukocyte chemotaxis 1, vascular leakage 2, endothelial cell migration (but not proliferation) 3, smooth muscle cells proliferation 4, and astrocytes proliferation 5, 6. In the human body, they are derived de novo from arachidonic acid 7 and include five types, namely leukotriene A4 (LTA4), leukotriene B4 (LTB4), leukotriene C4 (LTC4), leukotriene D4 (LTD4), and leukotriene E4 (LTE4). LTA4 and LTB4 (non‐cysteinyl leukotrienes) are structurally different from the cysteinyl leukotrienes (Cys‐LT) as they lack the cysteine moiety, which is present in the Cys‐LT (LTC4, LTD4, and LTE4) 8. Their hospitality is also welcomed by other type of receptors such as BLT1 and BLT2 9, whereas LTC4, LTD4, and LTE4 are the ligands majorly for cysteinyl leukotrienes type 1 (CysLT1R) and type 2 receptor (CysLT2R) 10. The rank of order is LTD4 > LTC4 > LTE4 by means of their affinity toward CysLT1R 11, whereas the order to that of CysT2R is LTC4 = LTD4 >> LTE4 12. Apart from these two main receptors, several other receptors have been reported but their role as Cys‐LT receptors is very little known. These additionally reported receptors are GPR17 13, GPR99 2, PPARγ 14, and P2Y12 15, 16. The latter three receptors, that is, GPR99, PPARγ, and P2Y12, have been reported to have preferential binding to LTE4, which, in general, has less preference for CysLT1R or CysLT2R (Figure 1). Structurally, CysLT1R and CysLT2R are 38% homologous to each other 12, and they also share a phylogenetic relationship with the purinergic (P2Y) class of GPCRs 15, 17 and receptors for proteases (PARs), and platelet‐activating factor receptor (PAFR) 18, 19. Apart from these, a cross talk between the toll‐like receptors (TLRs) and CysLT1R in dendritic cells has also been reported 20.

Figure 1.

Figure 1

Biosynthesis pathway of the Cys‐LT and their receptors. cPLA2‐derived arachidonic acid is converted to LTA4 by the action of 5‐LOX and FLAP (this step can be blocked by 5‐LOX/FLAP inhibitor zileuton). LTA4 is then rapidly converted to LTB4 by LTA4‐H or to LTC4 by LTC4‐S; LTC4 is further converted to LTD4 and LTE4. LTC4, LTD4, and LTE4 can bind to CysLT 1R, CysLT 2R, and GPR17 (CysLT 1R antagonists can block the activity of CysLT 1R or GPR17, but not CysLT 2R). Among the Cys‐LT, only LTE4 shows preferential binding toward GPR99, PPAR γ, and P2Y12.

Ever since the elucidation of chemical structure of the Cys‐LT and their association with inflammation 7, their role has been mainly studied in the pathophysiology of asthma 21, 22, 23. However, in more recent days, their role in other diseases has been emerging and pathophysiological role of these lipid mediators in diseases such as cardiovascular diseases including vascular inflammation and atherosclerosis 24, 25, 26, cancer 27, 28, 29, and many CNS diseases (discussed in the later parts of this review) has been shown. Herein, a summary has been made on the research findings on Cys‐LT and their receptors in different CNS diseases and the underlying mechanism by which they might play a crucial role in such mediation of the CNS diseases.

Synthesis of the Cys‐LT through Arachidonic Acid Metabolism

Free arachidonic acid (AA), a ubiquitous, 20‐carbon chain fatty acid 30, which is liberated from membrane phospholipids through calcium‐dependent cPLA2 (cytosolic phospholipase A2) in the cytosol 31, is metabolized enzymatically to eicosanoids through three major pathways, namely cytochrome P450, cyclooxygenase (COX), and lipoxygenase (LOX) pathways 28. In the cytochrome P450 pathway, AA is converted into epoxyeicosatrienoic acids (EETs), hydroxyeicosatetraenoic acids (HETEs), and hydroperoxyeicosatetraenoic acids (HPETEs). In the COX pathway, AA is metabolized to prostaglandin H2 (PGH2), which is further metabolized, by particular prostaglandin and thromboxane synthases, to prostanoids, together with prostaglandins (PGs), for example, PGE2, PGF2, PGD2 and PGI2 and thromboxanes (TXs), for example, TXA2. In the LOX pathway, AA is metabolized to 8‐, 12‐ and 15‐HPETE by 12‐ and 15‐LOX or to 5‐HPETE by 5‐LOX and 5‐lipoxygenase‐activating protein (FLAP). 5‐HPETE instantly undergoes dehydration to form LTA4, which is unstable in nature and rapidly metabolized either to produce LTB4 by the act of LTA4 hydrolase (LTA4‐H) or to generate LTC4 by the action of LTC4 synthase (LTC4‐S); LTC4 is further converted to LTD4 and LTE4 32 (Figure 1).

Although the biosynthesis process of the Cys‐LT mainly occurs in a cell‐specific manner in the nuclear envelope 33, a nonspecific mechanism may also be followed by cells like vascular endothelial cells 34, 35, and platelets 36 to produce Cys‐LT. Although these cells lack the enzymes to produce LTA4, they can use the LTA4 from the surrounding neutrophils and produce LTC4 ‐ an alternate route for Cys‐LT biosynthesis known as transcellular biosynthesis 33. Such transcellular biosynthesis has also been observed in neuronal and glial cells when these cells are cocultured with neutrophils 37; similar trend was found in brain endothelial cells in a CysLT2R‐dependent manner 38. Neutrophils infiltrate the brain regions following injury and donate LTA4 to these cells, which is further utilized to generate Cys‐LT in a similar manner to that of followed in the traditional Cys‐LT biosynthesis (Figure 2).

Figure 2.

Figure 2

Transcellular biosynthesis of Cys‐LT in the brain. Neutrophils, which infiltrate the inflamed brain area, transfer LTA4 to the surrounding neurons, glial cells, and brain endothelial cells. This LTA4 then can further be utilized by these brain resident cells to produce Cys‐LT.

Receptors for the Cys‐LT

The main two receptors for Cys‐LT, namely CysLT1R and CysLT2R, are the members of G‐protein‐coupled receptors (GPCRs) 39, 40. Recombinant CysLTRs activate the Gq/11 pathway that involves modulation of inositol phospholipids hydrolysis and calcium mobilization, whereas the native systems involve activation of a pertussis toxin‐insensitive Gi/o‐protein 39. The internalization and signaling of CysLT1R, as a GPCR, largely depends on protein kinase C (PKC) 41, 42, which is the principal regulator of both rapid agonist‐dependent internalization and rapid agonist‐dependent desensitization of the receptor 41. No data are yet available on the functional regulation of CysLT2R 39.

CysLT1R (Cysteinyl Leukotrienes Type 1 Receptor)

The first known receptor of the Cys‐LT, CysLT1R, was reported in 1999 11, 43. The receptor showed highest affinity toward LTD4 (EC50 = 2.5 nM), with much lower affinity toward LTC4 (EC50 = 24 nM) and LTE4 (EC50 = 240 nM) 43. The human CysLT1R shares a homology of 38% with CysLT2R 12, 32% with the purinergic receptor P2Y1 and the receptor for platelet‐activating factor, and 28% with BLT receptor 11. The receptor contains 336 amino acid residues with found mRNA expression in the spleen, lung tissue, smooth muscle cells, leukocytes, macrophages, and mast cells 10, 33. Later interpretation in mouse led to the discovery that the mCysLT1R was composed of 339 amino acid residues with 87.3% identity with the hCysLT1R 44. The receptor is the primary target in the treatment of asthma and can be targeted by agents such as montelukast, pranlukast, and zafirlukast.

CysLT2R (Cysteinyl Leukotrienes Type 2 Receptor)

One year after CysLT1R was cloned, another receptor for the Cys‐LT was reported in 2000. The receptor was named CysLT2R that showed an equal affinity both toward LTC4 and LTD4 with a lower affinity toward LTE4 12, 45. More recently, it has been confirmed that LTC4 selectively binds to CysLT2R and works on a CysLT2R‐dependant manner and that LTC4 is inactivated in CysLT2R−/− mice 46. The receptor contains 345 amino acid residues and is expressed in the adrenal gland, heart, spleen, leukocytes, placenta, brain, and lymph nodes, with lower expression in the lung 10, 33. Later studies carried out on mCysLT2R found that the homology of mCysLT2R to that of hCysLT2R was 73.4% and it consisted of 309 amino acids 47. It is interesting that CysLT2R uses different activation niche for the mediation of its inflammatory responses 48.

GPR17 (G‐protein‐coupled Receptor 17)

GPR17, a receptor with close phylogenetic relationship with the purinergic receptors 49, is a G‐coupled dual receptor activated by both uracil nucleotides and Cys‐LT 13, and LTD4 can induce migratory function of the receptor 50. Whereas several studies support the idea that GPR17 is a receptor for Cys‐LTs, other studies also exist contradicting the same. One study represented that GPR17 is not a cognate receptor for Cys‐LT 51, and rather, it acts a negative regulator of CysLT1R 52.

GPR99 (G‐protein‐coupled Receptor 99)

GPR99, another GPCR belonging to the P2Y subfamily of GPCRs 53, was recently identified to be a receptor for LTE4 2. LTE4, which generally has the lowest affinity toward CysLT1R and CysLT2R, was shown to be involved in vascular leakage through GPR99‐mediated actions 2.

PPARγ (Peroxisome Proliferator‐activated Receptor γ)

Although LTB4, the most important subtype of the noncysteinyl leukotrienes, is known as an important regulator of the functions of the PPAR family of receptors 54, only a single study exists representing PPARγ as a receptor for the Cys‐LT 14. Paruchuri and colleagues, in 2008, using culture of human mast cells, showed that PPARγ is involved in LTE4‐mediated ERK (Extracellular signal‐Regulated Kinases) activation and that treatment with GW9622, a selective PPARγ antagonist, can block the LTE4‐induced, but not LTD4‐induced, activation of ERK, suggesting a selective binding of LTE4 with PPARγ 14.

P2Y12

P2Y12 was shown as a receptor for LTE4 15, 16. However, a recent study has opposed that LTE4 or other Cys‐LT do not bind to P2Y12 55. On the contrary, again, even a more recent study showed LTE4 as a ligand for the P2Y12 56, suggesting a need for more studies confirming the crosstalk between LTE4/P2Y12. P2Y12 has also been recognized as to support the cross talk between LTC4 and CysLT2R 46.

P2Y6 (A Supporting Receptor)

Although this purinergic subfamily of receptor does not bind to the Cys‐LT, the activity of the Cys‐LT/their receptors depends on these receptors. P2Y6 has been identified as a mediator of chemokine generation and promoter of cell survival that supports the activity of CysLT1R 57. CysLT1R antagonists also show differential inhibitory effect on the P2Y6 receptor 58.

Therapeutic Interventions: Cys‐LT Biosynthesis Blockers and the CysLTR Antagonists

Currently, there are two available approaches to therapeutically block the Cys‐LT. The 5‐LOX pathway of Cys‐LT production during AA metabolism can be blocked by FLAP inhibitors such as zileuton, which is a choice of drug in asthma. Drugs targeting CysLT1R such as montelukast, pranlukast, and zafirlukast are also in use for asthma and allergic rhinitis and are well tolerated with low therapeutic‐to‐toxic ratio 33, 59, 60. These drugs mediate their effect by selective antagonism of CysLT1R and have no effect on CysLT2R. Although there are no clinically available CysLT2R antagonists, development of such agents is deemed necessary as some patients fail to respond to CysLT1R antagonists 33, 61. An agent was developed as a dual antagonist of CysLT1R and CysLT2R, named BAYu9773, but it showed poor potency and selectivity for these receptors in human tissues 62. More dual antagonists are also under development 63.

5‐LOX/Cys‐LT/CysLTRs in CNS Diseases

Although 5‐LOX, Cys‐LT, and CysLTRs are traditionally known targets in asthma and other allergic diseases, their role also has been shown to be associated with other human diseases such as in cancer, atherosclerosis, and several CNS diseases. Among the CNS diseases, they are related to brain injury, Multiple sclerosis, Alzheimer's disease, Parkinson's disease, Huntington's disease, epilepsy, and depression and they also facilitate viral entry into the brain (Figure 3). Although normal physiological role of 5‐LOX in the brain remains elusive, its pathophysiological role is more extensively studied 8. 5‐LOX is expressed on neurons and regulates synaptic plasticity, and brain aging 64, 65. Age‐dependent higher expression of 5‐LOX is seen in aged mouse, rat, and human hippocampus 65, 66, and glucocorticoid receptor‐dependent mechanism might be one of the regulatory mechanisms involved in neuronal expression of 5‐LOX 67. Age‐related increased expression of CysLT1R has been also shown in aged rat brain, localized in glial cells and several types of neuronal cells 66. While the basal expression of the CysLTRs, especially CysLT1R 43, is very weak in the normal brain, it remains unclear how the receptor is upregulated in the inflamed brain. Although acute inflammation relative of the CysLT1R signaling is important in reparative responses in injured brain 68, chronic inflammation, however, is considered harmful in such scenario 69. Whereas glial cells, namely microglia and astrocytes, are regarded as the key players in neuroinflammation, their activity has been shown to be regulated by Cys‐LT and CysLT1R. For instance, LTD4 activates mouse microglial cells in vitro, which is mediated by CysLT1R 70. Microglia are the resident immune cells in the brain, which are activated following brain injury, and their activation can either be harmful (M1 pathway) or be beneficial (M2 pathway) 71. Our recent study also indicates the role of CysLT1R in bacterial lipopolysaccharide‐induced neuroinflammation in mice; we downregulated the activity of the receptor using its shRNA‐mediated knockdown and pharmacological inhibition by pranlukast to study the role of the receptor in microglial activation, and we found that such activation of microglia was normalized in CysLT1R‐shRNA‐ or pranlukast‐treated animal groups along with the downregulation of the M1 pathway molecules such as IL‐1β, and TNF‐α (F. Chen, A. Ghosh, F. Wu, S. Tang, M. Hu, H. Sun, L. Kong and H. Hong, unpublished data). Another study has depicted the role of 5‐LOX, and LTD4 in TGF‐β1‐induced astrocyte migration through the activation of CysLT1R 72. Administration of zileuton or montelukast attenuated such migration of astrocytes, suggesting a role of these molecules in astrocyte‐mediated neuroinflammation. However, in the same study, administration of a CysLT2R‐specific antagonist had no effect on such phenomenon, suggesting a greater role of the CysLT1R rather than the CysLT2R in neuroinflammation.

Figure 3.

Figure 3

Contribution of 5‐LOX/Cys‐LT/CysLTRs in CNS and other diseases. They are traditionally known for mediation of asthma and the CysLT 1R antagonists or 5‐LOX inhibitors are used for clinical management of the disease; montelukast is also approved by the Food and Drug Administration of the USA for the management of other allergic diseases. However, the importance of these inflammatory molecules is emerging in other fields and has been shown to be associated with cancer, atherosclerosis, and several brain diseases as displayed in the Figure. (Figure illustrations were created using MOTIFOLIO Biomedical PowerPoint Toolkits for Presentations and Microsoft Office PowerPoint).

Brain Injury

Association of Cys‐LT/their receptors has been shown in different types of brain injury, including traumatic brain injury 73, 74, ischemia‐related brain injury 75, 76, 77, and brain cryoinjury 78, 79.

However, the concentration of free arachidonic acid is usually very low in the brain, but it can increase greatly following different stimuli such as ischemia 80. Cerebral ischemia is clinically characterized by reduced cerebral hypoperfusion, which causes reduced blood circulation, wholly or partly, to the parts of the brain and accompanied by neuroinflammation and neuronal death. In global ischemic models, an augmented formation of Cys‐LT has been demonstrated following reperfusion 81, 82, 83. Moreover, higher level of these lipid mediators is also seen in cerebrospinal fluid (CSF) of patients with acute cerebral ischemia 84. Although the role of LTs in the developmental process of trauma‐associated ischemia is not well known, interestingly, cPLA2‐deficient mice are less susceptible to cerebral ischemia/reperfusion injury 85, 86. The first evidence of neuroprotection, through postischemic modulation of LT levels, was obtained by the use of MK‐886, an LT synthesis inhibitor, in an in vivo model of pMCAO (permanent occlusion of the middle cerebral artery) in rats 87. Whereas neuroinflammation is a critical component following brain injury, it is accompanied by an aggravated level of Cys‐LT receptors 88. Despite the fact that CysLT2R is the main isoform of CysLTRs in the normal brain, the first line of data, from experiments carried out with CysLTR antagonists, suggested that selective CysLT1R antagonists, including pranlukast and montelukast, might have a protective effect in focal cerebral ischemia 89, 90; protective effect of montelukast against global ischemia was also shown 91. However, recent studies showed spatiotemporal expression of CysLT2R in cerebral ischemia 75 and that using HAMI 3379, a CysLT2 receptor antagonist, is neuroprotective against ischemic injury and neuroinflammation 76, 92. Association of GPR17 in ischemia‐related neuroinflammation has also been shown 93. The neuroprotective effect of the FLAP inhibitor zileuton and genetic disruption of ALOX5AP has also been shown to ameliorate ischemic stroke and reduce infarct size and neuroinflammation following cerebral ischemia 94, 95, 96. Moreover, genetic association studies have linked the risk of ischemic stroke with the leukotrienes biosynthesis pathway 97, 98, 99.

Multiple Sclerosis/Experimental Autoimmune Encephalomyelitis

Multiple sclerosis (MS) is a severe neurological disease characterized by autoimmunity‐mediated demyelination, oligodendrocyte damage, and, ultimately, axonal loss 100. Despite an increasing appreciation of the importance of remyelination, most therapeutic approaches for MS are immunomodulatory drugs that target the inflammatory component of the disease 49. Increased expression of 5‐LOX in lesions 101, 102 and of 5‐LOX‐derived LT products in the cerebrospinal fluid 103, 104 is found in patients with MS. Yoshikawa and colleagues showed that pharmacological inhibition of 5‐LOX could attenuate axonal damage and motor deficits related to MS pathology 105. Demyelination of the CNS relative of arachidonic acid cascade was also suggested by studies in models of experimental autoimmune encephalomyelitis (EAE) 106, 107. Moreover, the effector phase of EAE can be ameliorated by targeting cPLA(2)α, which is the precursor of the LTs 108. Infiltration of Th17 cells in the inflamed area of the brain, which is a characteristic of EAE, can be blocked by inhibiting LTs 109, and inhibition of Th1/Th17 by the FLAP inhibitor zileuton and Cys‐LT antagonists is fruitful in a model of EAE 110, 111. The role of GPR17 and purinergic signaling has also been strongly suggested as a reparative approach in MS 49, 112.

Alzheimer's Disease

Alzheimer's disease (AD) is the most prevalent form of dementia, pathologically characterized by extracellular senile plaques consisting of β‐amyloid (Aβ) plaques and intracellular neurofibrillary tangles (NFTs) of tau protein owing to a progressive loss of memory and cognition. Using different in vivo and in vitro studies, it has been shown that LTD4‐induced upregulation of CysLT1R is correlated with increased Aβ, and amyloid precursor protein (APP) and is associated with cognitive dysfunctions in mice 113, 114, 115; such pathological symptoms are attenuated through administration of selective CysLT1R antagonists such as pranlukast and montelukast 116, 117. Moreover, the CysLT1R antagonists mediate structural and functional rejuvenation of the aged brain through several mechanisms such as by downregulating GPR17 and CysLT1R, reducing microglial reactivity, and by maintaining blood–brain barrier integrity 66. 5‐LOX and FLAP have also long been recognized to be associated with different forms of AD 118, 119, 120, 121 with several mechanisms 122, 123, 124. 5‐LOX is elevated in AD brain and is immunoreactive that might be involved in posttranslational modifications of the Aβ senile plaques and NFTs 125. It is an endogenous modulator of Aβ formation in vivo 126 and participates in corticosteroid‐dependent Aβ generation 127. In line with the fact that Aβ aggregation is dependent of 5‐LOX, agents as dual inhibitors of Aβ and 5‐LOX have been developed 128. Pharmacological studies using zileuton also exist showing ameliorative effect of the drug on AD phenotypes in different animal models 129, 130, 131. Genetic knockout study on the 5‐LOX gene has also evidenced similar beneficial effects against AD pathology supporting the pharmacological findings 127.

While the aforementioned studies mainly focused on the familial form of AD, the sporadic form of AD is also important to consider. Whereas the incidence of sporadic AD is largely characterized by oxidative stress, neuroinflammation and a great load of proinflammatory cytokines, the 5‐LOX pathway regulates the proinflammatory mediators in the cerebral cortex 132. COX/5‐LOX are mediators of such inflammation‐related neurotoxicity 133 and licofelone, a novel dual inhibitor of COX/5‐LOX, reduces oxidative stress and increased burden of proinflammatory cytokines in a rat model of sporadic AD 134.

Mild cognitive impairment (MCI), which serves as a prodrome to AD, is also associated with 5‐LOX upregulation in the early development of the disease 135, and montelukast shows protective effect in an animal model of MCI 136.

Parkinson's Disease

Parkinson's disease (PD) is pathologically characterized by degeneration of the dopaminergic neurons in the substantia nigra of the brain and manifested by the movement disorders in elderly populations. The LOX isozymes, including 5‐LOX, are involved in the maintenance of normal dopaminergic function in the striatum and the differentially contribute to striatal vulnerability in response to neurotoxicity 137. Another study highlighted the participation of the 5‐LOX system in MPTP‐induced PD model 138; however, the activity of LTB4 mainly was shown in such mediation of the disease rather than the Cys‐LT. Wang et al. 139 studied the expression pattern of the CysLT1R, CysLT2R, and GPR17 in PD mouse model. Although the expression of CysLT1R and GPR17 was found to be decreasing, CysLT2R was shown to be increasing and expressed mainly in the proliferating astrocytes. Interestingly, CysLT2R is the mediator of inflammation and consequent neurotoxicity in vitro PD model 140.

Epilepsy

Epilepsy‐related BBB (blood–brain barrier) disruption has been shown to be mediated by Cys‐LT 141, and modulation of Cys‐LT by montelukast suppresses the development and frequency of seizures in vivo 142, 143. Clinical evidence also exists highlighting the efficacy of pranlukast in epileptic patients 144.

Viral Entry into the Brain

Cys‐LT and other AA metabolites have been shown to facilitate pathogen entry into the brain through disruption of the BBB. Bertin et al. 145 have shown that Cys‐LT can facilitate the entry of HIV‐1 into the brain, which contributes to HIV‐1‐mediated CNS dysfunctions through the CX3CL1/fractalkine‐mediated transmigration of the virus through infected CD4+ T cells. Another study has depicted the relevance of these inflammatory molecules in Escherichia coli‐mediated meningitis 146. PKCα, which controls the activity of the Cys‐LT in many pathophysiological processes, is also a regulator of the activity of the Cys‐LT during such viral invasion across the BBB, according to the mechanisms reported in this study 146. In the same study, blockade of the activity of cPLA2 α and 5‐LOX, the precursors of arachidonic acid and Cys‐LT synthesis, respectively, recovered the incidence of viral entry into the brain suggesting a greater role of the Cys‐LT and arachidonic acid metabolism pathways in facilitating viral entry into the brain. Higher expression of 5‐LOX was also found in dolphin brains with encephalitis 147.

Others

Limited evidence has shown the efficacy of montelukast in an animal model of Huntington's disease (HD) 148. Montelukast, given orally, ameliorated mitochondrial dysfunction and TNF‐α level relative of the disease pathology; however, the study did not show whether montelukast mediated such effect by downregulation of Cys‐LT/their receptors. Another study exists depicting the role of CysLT1R in depressive behavior and neuroinflammation in a mouse model of chronic mild stress (CMS) 149. The study showed that hippocampal expression of CysLT1R is increased in the CMS‐exposed mice in a time‐dependent manner and lentivirus‐mediated knockdown of the receptor in the mouse hippocampus prevents such CMS‐induced depressive behaviors.

Conclusive Remarks and Future Outlook

The inflammatory milieu relative of Cys‐LT has been implicated in a range of pathological conditions such as in asthma, allergic rhinitis, cardiovascular disease, cancer, and neurological dysfunctions. Although many aspects of the roles of the Cys‐LT and their receptors, as mediators of inflammation, in asthma and other allergic diseases are well established, their pathophysiological role in the brain is very little known and much more attention should be given to gain the utmost understanding of their role in CNS diseases.

Conflict of Interest

The authors declare no conflict of interest.

Acknowledgments

The work is supported by grants from the Natural Science Foundation of China (81273497 and 81573413 to HH) and the Natural Science Foundation of Jiangsu Province (SBK201320969 to HH). AG is a recipient of China Govt. Scholarship‐Chinese University Program (China Scholarship Council) for foreign students at CPU. FC is a recipient of PhD Research Assistantship at CPU. AT is a recipient of Postgraduate Scholarship at CityU HK.

References

  • 1. Bautz F, Denzlinger C, Kanz L, et al. Chemotaxis and transendothelial migration of CD34(+) hematopoietic progenitor cells induced by the inflammatory mediator leukotriene D4 are mediated by the 7‐transmembrane receptor CysLT1. Blood 2001;97:3433–4340. [DOI] [PubMed] [Google Scholar]
  • 2. Kanaoka Y, Maekawa A, Austen KF. Identification of GPR99 protein as a potential third cysteinyl leukotriene receptor with a preference for leukotriene E4 ligand. J Biol Chem 2013;288:10967–10972. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Yuan YM, Fang SH, Qian XD, et al. Leukotriene D4 stimulates the migration but not proliferation of endothelial cells mediated by the cysteinyl leukotriene cysLT1 receptor via the extracellular signal‐regulated kinase pathway. J Pharmacol Sci 2009;109:285–292. [DOI] [PubMed] [Google Scholar]
  • 4. Ravasi S, Citro S, Viviani B, et al. CysLT1 receptor‐induced human airway smooth muscle cells proliferation requires ROS generation, EGF receptor transactivation and ERK1/2 phosphorylation. Respir Res 2006;7:42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Ciccarelli R, D'Alimonte I, Santavenere C, et al. Cysteinyl‐leukotrienes are released from astrocytes and increase astrocyte proliferation and glial fibrillary acidic protein via cys‐LT1 receptors and mitogen‐activated protein kinase pathway. Eur J Neurosci 2004;20:1514–1524. [DOI] [PubMed] [Google Scholar]
  • 6. Huang X‐J, Zhang W‐P, Li C‐T, et al. Activation of CysLT receptors induces astrocyte proliferation and death after oxygen–glucose deprivation. Glia 2008;56:27–37. [DOI] [PubMed] [Google Scholar]
  • 7. Samuelsson B. Leukotrienes: Mediators of immediate hypersensitivity reactions and inflammation. Science 1983;220:568–575. [DOI] [PubMed] [Google Scholar]
  • 8. Kuhn H, Banthiya S, van Leyen K. Mammalian lipoxygenases and their biological relevance. Biochim Biophys Acta 2015;1851:308–330. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Bäck M, Powell WS, Dahlén S‐E, et al. Update on leukotriene, lipoxin and oxoeicosanoid receptors: IUPHAR Review 7. Brit J Pharmacol 2014;171:3551–3574. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Haeggstrom JZ, Funk CD. Lipoxygenase and leukotriene pathways: Biochemistry, biology, and roles in disease. Chem Rev 2011;111:5866–5898. [DOI] [PubMed] [Google Scholar]
  • 11. Lynch KR, O'Neill GP, Liu Q, et al. Characterization of the human cysteinyl leukotriene CysLT1 receptor. Nature 1999;399:789–793. [DOI] [PubMed] [Google Scholar]
  • 12. Heise CE, O'Dowd BF, Figueroa DJ, et al. Characterization of the human cysteinyl leukotriene 2 receptor. J Biol Chem 2000;275:30531–30536. [DOI] [PubMed] [Google Scholar]
  • 13. Ciana P, Fumagalli M, Trincavelli ML, et al. The orphan receptor GPR17 identified as a new dual uracil nucleotides/cysteinyl‐leukotrienes receptor. EMBO J 2006;25:4615–4627. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Paruchuri S, Jiang Y, Feng C, et al. Leukotriene E4 activates peroxisome proliferator‐activated receptor gamma and induces prostaglandin D2 generation by human mast cells. J Biol Chem 2008;283:16477–16487. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Nonaka Y, Hiramoto T, Fujita N. Identification of endogenous surrogate ligands for human P2Y12 receptors by in silico and in vitro methods. Biochem Biophys Res Commun 2005;337:281–288. [DOI] [PubMed] [Google Scholar]
  • 16. Paruchuri S, Tashimo H, Feng C, et al. Leukotriene E4–induced pulmonary inflammation is mediated by the P2Y12 receptor. J Exp Med 2009;206:2543–2555. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Mellor EA, Maekawa A, Austen KF, et al. Cysteinyl leukotriene receptor 1 is also a pyrimidinergic receptor and is expressed by human mast cells. Proc Natl Acad Sci USA 2001;98:7964–7969. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Fredriksson R, Lagerstrom MC, Lundin LG, et al. The G‐protein‐coupled receptors in the human genome form five main families. Phylogenetic analysis, paralogon groups, and fingerprints. Mol Pharmacol 2003;63:1256–1272. [DOI] [PubMed] [Google Scholar]
  • 19. Kroeze WK, Sheffler DJ, Roth BL. G‐protein‐coupled receptors at a glance. J Cell Sci 2003;116:4867–4869. [DOI] [PubMed] [Google Scholar]
  • 20. Thivierge M, Stankova J, Rola‐Pleszczynski M. Toll‐like receptor agonists differentially regulate cysteinyl‐leukotriene receptor 1 expression and function in human dendritic cells. J Allergy Clin Immunol 2006;117:1155–1162. [DOI] [PubMed] [Google Scholar]
  • 21. Drazen JM. Leukotrienes in asthma. Adv Exp Med Biol 2003;525:1–5. [DOI] [PubMed] [Google Scholar]
  • 22. Hallstrand TS, Henderson WR Jr. An update on the role of leukotrienes in asthma. Curr Opin Allergy Clin Immunol 2010;10:60–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Laidlaw TM, Boyce JA. Cysteinyl leukotriene receptors, old and new; implications for asthma. Clin Exp Allergy 2012;42:1313–1320. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Back M, Hansson GK. Leukotriene receptors in atherosclerosis. Ann Med 2006;38:493–502. [DOI] [PubMed] [Google Scholar]
  • 25. Back M. Leukotriene receptors: Crucial components in vascular inflammation. Sci World J 2007;7:1422–1439. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Ingelsson E, Yin L, Back M. Nationwide cohort study of the leukotriene receptor antagonist montelukast and incident or recurrent cardiovascular disease. J Allergy Clin Immunol 2012;129:702–707. [DOI] [PubMed] [Google Scholar]
  • 27. Savari S, Vinnakota K, Zhang Y, et al. Cysteinyl leukotrienes and their receptors: Bridging inflammation and colorectal cancer. World J Gastroenterol 2014;20:968–977. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Cathcart M‐C, Lysaght J, Pidgeon GP. Eicosanoid signalling pathways in the development and progression of colorectal cancer: Novel approaches for prevention/intervention. Cancer Metastasis Rev 2011;30:363–385. [DOI] [PubMed] [Google Scholar]
  • 29. Matsuyama M, Funao K, Hayama T, et al. Relationship between cysteinyl‐leukotriene‐1 receptor and human transitional cell carcinoma in bladder. Urology 2009;73:916–921. [DOI] [PubMed] [Google Scholar]
  • 30. Drazen JM, Austen KF. Leukotrienes and airway responses. Am Rev Respir Dis 1987;136:985–998. [DOI] [PubMed] [Google Scholar]
  • 31. Clark JD, Lin LL, Kriz RW, et al. A novel arachidonic acid‐selective cytosolic PLA2 contains a Ca(2+)‐dependent translocation domain with homology to PKC and GAP. Cell 1991;65:1043–1051. [DOI] [PubMed] [Google Scholar]
  • 32. Drazen JM, Israel E, O'Byrne PM. Treatment of Asthma with Drugs Modifying the Leukotriene Pathway. N Engl J Med 1999;340:197–206. [DOI] [PubMed] [Google Scholar]
  • 33. Capra V, Thompson MD, Sala A, et al. Cysteinyl‐leukotrienes and their receptors in asthma and other inflammatory diseases: Critical update and emerging trends. Med Res Rev 2007;27:469–527. [DOI] [PubMed] [Google Scholar]
  • 34. Feinmark SJ, Cannon PJ. Endothelial cell leukotriene C4 synthesis results from intercellular transfer of leukotriene A4 synthesized by polymorphonuclear leukocytes. J Biol Chem 1986;261:16466–16472. [PubMed] [Google Scholar]
  • 35. Maclouf J, Murphy RC, Henson PM. Transcellular sulfidopeptide leukotriene biosynthetic capacity of vascular cells. Blood 1989;74:703–707. [PubMed] [Google Scholar]
  • 36. Maclouf JA, Murphy RC. Transcellular metabolism of neutrophil‐derived leukotriene A4 by human platelets. A potential cellular source of leukotriene C4. J Biol Chem 1988;263:174–181. [PubMed] [Google Scholar]
  • 37. Farias SE, Zarini S, Precht T, et al. Transcellular biosynthesis of cysteinyl leukotrienes in rat neuronal and glial cells. J Neurochem 2007;103:1310–1318. [DOI] [PubMed] [Google Scholar]
  • 38. Di Gennaro A, Carnini C, Buccellati C, et al. Cysteinyl‐leukotrienes receptor activation in brain inflammatory reactions and cerebral edema formation: A role for transcellular biosynthesis of cysteinyl‐leukotrienes. FASEB J 2004;18:842–844. [DOI] [PubMed] [Google Scholar]
  • 39. Rovati GE, Capra V. Cysteinyl‐leukotriene receptors and cellular signals. Sci World J 2007;7:1375–1392. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Hedi H, Norbert G. 5‐lipoxygenase pathway, dendritic cells, and adaptive immunity. J Biomed Biotechnol 2004;2004:99–105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Naik S, Billington CK, Pascual RM, et al. Regulation of cysteinyl leukotriene type 1 receptor internalization and signaling. J Biol Chem 2005;280:8722–8732. [DOI] [PubMed] [Google Scholar]
  • 42. Deshpande DA, Pascual RM, Wang SW, et al. PKC‐dependent regulation of the receptor locus dominates functional consequences of cysteinyl leukotriene type 1 receptor activation. FASEB J 2007;21:2335–2342. [DOI] [PubMed] [Google Scholar]
  • 43. Sarau HM, Ames RS, Chambers J, et al. Identification, molecular cloning, expression, and characterization of a cysteinyl leukotriene receptor. Mol Pharmacol 1999;56:657–663. [DOI] [PubMed] [Google Scholar]
  • 44. Ogasawara H, Ishii S, Yokomizo T, et al. Characterization of mouse cysteinyl leukotriene receptors mCysLT1 and mCysLT2: Differential pharmacological properties and tissue distribution. J Biol Chem 2002;277:18763–18768. [DOI] [PubMed] [Google Scholar]
  • 45. Nothacker HP, Wang Z, Zhu Y, et al. Molecular cloning and characterization of a second human cysteinyl leukotriene receptor: Discovery of a subtype selective agonist. Mol Pharmacol 2000;58:1601–1608. [DOI] [PubMed] [Google Scholar]
  • 46. Cummings HE, Liu T, Feng C, et al. Cutting edge: Leukotriene C4 activates mouse platelets in plasma exclusively through the type 2 cysteinyl leukotriene receptor. J Immunol 2013;191:5807–5810. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Hui Y, Yang G, Galczenski H, et al. The murine cysteinyl leukotriene 2 (CysLT2) receptor. cDNA and genomic cloning, alternative splicing, and in vitro characterization. J Biol Chem 2001;276:47489–47495. [DOI] [PubMed] [Google Scholar]
  • 48. Ni NC, Ballantyne LL, Mewburn JD, et al. Multiple‐site activation of the cysteinyl leukotriene receptor 2 is required for exacerbation of ischemia/reperfusion injury. Arterioscler Thromb Vasc Biol 2014;34:321–330. [DOI] [PubMed] [Google Scholar]
  • 49. Hennen S, Wang H, Peters L, et al. Decoding signaling and function of the orphan G protein‐coupled receptor GPR17 with a small‐molecule agonist. Sci Signal 2013;6:ra93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Cosentino S, Castiglioni L, Colazzo F, et al. Expression of dual Nucleotides/Cysteinyl‐Leukotrienes Receptor GPR17 in early trafficking of cardiac stromal cells after myocardial infarction. J Cell Mol Med 2014;18:1785–1796. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Qi AD, Harden TK, Nicholas RA. Is GPR17 a P2Y/leukotriene receptor? examination of uracil nucleotides, nucleotide sugars, and cysteinyl leukotrienes as agonists of GPR17. J Pharmacol Exp Ther 2013;347:38–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Maekawa A, Balestrieri B, Austen KF, et al. GPR17 is a negative regulator of the cysteinyl leukotriene 1 receptor response to leukotriene D4. Proc Natl Acad Sci USA 2009;106:11685–11690. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Wittenberger T, Hellebrand S, Munck A, et al. GPR99, a new G protein‐coupled receptor with homology to a new subgroup of nucleotide receptors. BMC Genom 2002;3:17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. Marion‐Letellier R, Savoye G, Ghosh S. Fatty acids, eicosanoids and PPAR gamma. Eur J Pharmacol In press. DOI: 10.1016/j.ejphar.2015.11.004. [DOI] [PubMed] [Google Scholar]
  • 55. Foster HR, Fuerst E, Lee TH, et al. Characterisation of P2Y(12) receptor responsiveness to cysteinyl leukotrienes. PLoS ONE 2013;8:e58305. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Shirasaki H, Kanaizumi E, Seki N, et al. Leukotriene E4 induces MUC5AC release from human airway epithelial NCI‐H292 cells. Allergol Int 2015;64:169–174. [DOI] [PubMed] [Google Scholar]
  • 57. Jiang Y, Borrelli L, Bacskai BJ, et al. P2Y6 receptors require an intact cysteinyl leukotriene synthetic and signaling system to induce survival and activation of mast cells. J Immunol 2009;182:1129–1137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Lau WK, Chow AW, Au SC, et al. Differential inhibitory effects of CysLT(1) receptor antagonists on P2Y(6) receptor‐mediated signaling and ion transport in human bronchial epithelia. PLoS ONE 2011;6:e22363. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Keam SJ, Lyseng‐Williamson KA, Goa KL. Pranlukast: A review of its use in the management of asthma. Drugs 2003;63:991–1019. [DOI] [PubMed] [Google Scholar]
  • 60. Scott JP, Peters‐Golden M. Antileukotriene agents for the treatment of lung disease. Am J Respir Crit Care Med 2013;188:538–544. [DOI] [PubMed] [Google Scholar]
  • 61. Drazen JM, Silverman EK, Lee TH. Heterogeneity of therapeutic responses in asthma. Brit Med Bull 2000;56:1054–1070. [DOI] [PubMed] [Google Scholar]
  • 62. Labat C, Ortiz JL, Norel X, et al. A second cysteinyl leukotriene receptor in human lung. J Pharmacol Exp Ther 1992;263:800–805. [PubMed] [Google Scholar]
  • 63. Itadani S, Takahashi S, Ima M, et al. Discovery of a potent, orally available dual CysLT(1) and CysLT(2) antagonist with dicarboxylic acid. Bioorg Med Chem 2015;23:2079–2097. [DOI] [PubMed] [Google Scholar]
  • 64. Phillis JW, Horrocks LA, Farooqui AA. Cyclooxygenases, lipoxygenases, and epoxygenases in CNS: Their role and involvement in neurological disorders. Brain Res Rev 2006;52:201–243. [DOI] [PubMed] [Google Scholar]
  • 65. Chinnici CM, Yao Y, Praticò D. The 5‐lipoxygenase enzymatic pathway in the mouse brain: Young versus old. Neurobiol Aging 2007;28:1457–1462. [DOI] [PubMed] [Google Scholar]
  • 66. Marschallinger J, Schaffner I, Klein B, et al. Structural and functional rejuvenation of the aged brain by an approved anti‐asthmatic drug. Nat Commun 2015;6:8466. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Uz T, Dwivedi Y, Qeli A, et al. Glucocorticoid receptors are required for up‐regulation of neuronal 5‐lipoxygenase (5LOX) expression by dexamethasone. FASEB J 2001;15:1792–1794. [DOI] [PubMed] [Google Scholar]
  • 68. Kyritsis N, Kizil C, Zocher S, et al. Acute inflammation initiates the regenerative response in the adult zebrafish brain. Science 2012;338:1353–1356. [DOI] [PubMed] [Google Scholar]
  • 69. Akiyama H, Barger S, Barnum S, et al. Inflammation and Alzheimer's disease. Neurobiol Aging 2000;21:383–421. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. Yu SY, Zhang XY, Wang XR, et al. Cysteinyl leukotriene receptor 1 mediates LTD4‐induced activation of mouse microglial cells in vitro. Acta Pharmacol Sin 2014;35:33–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Graeber MB, Streit WJ. Microglia: Biology and pathology. Acta Neuropathol 2010;119:89–105. [DOI] [PubMed] [Google Scholar]
  • 72. Huang XQ, Zhang XY, Wang XR, et al. Transforming growth factor beta1‐induced astrocyte migration is mediated in part by activating 5‐lipoxygenase and cysteinyl leukotriene receptor 1. J Neuroinflammation 2012;9:145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Farias S, Frey LC, Murphy RC, et al. Injury‐related production of cysteinyl leukotrienes contributes to brain damage following experimental traumatic brain injury. J Neurotrauma 2009;26:1977–1986. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Corser‐Jensen CE, Goodell DJ, Freund RK, et al. Blocking leukotriene synthesis attenuates the pathophysiology of traumatic brain injury and associated cognitive deficits. Exp Neurol 2014;256:7–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Zhao CZ, Zhao B, Zhang XY, et al. Cysteinyl leukotriene receptor 2 is spatiotemporally involved in neuron injury, astrocytosis and microgliosis after focal cerebral ischemia in rats. Neuroscience 2011;189:1–11. [DOI] [PubMed] [Google Scholar]
  • 76. Zhang XY, Wang XR, Xu DM, et al. HAMI 3379, a CysLT2 receptor antagonist, attenuates ischemia‐like neuronal injury by inhibiting microglial activation. J Pharmacol Exp Ther 2013;346:328–341. [DOI] [PubMed] [Google Scholar]
  • 77. Zhao R, Shi W‐Z, Zhang Y‐M, et al. Montelukast, a cysteinyl leukotriene receptor‐1 antagonist, attenuates chronic brain injury after focal cerebral ischaemia in mice and rats. J Pharm Pharmacol 2011;63:550–557. [DOI] [PubMed] [Google Scholar]
  • 78. Qian X‐D, Wei E‐Q, Zhang L, et al. Pranlukast, a cysteinyl leukotriene receptor 1 antagonist, protects mice against brain cold injury. Eur J Pharmacol 2006;549:35–40. [DOI] [PubMed] [Google Scholar]
  • 79. Ding Q, Fang SH, Zhou Y, et al. Cysteinyl leukotriene receptor 1 partially mediates brain cryoinjury in mice. Acta Pharmacol Sin 2007;28:945–952. [DOI] [PubMed] [Google Scholar]
  • 80. Bosisio E, Galli C, Galli G, et al. Correlation between release of free arachidonic acid and prostaglandin formation in brain cortex and cerebellum. Prostaglandins 1976;11:773–781. [DOI] [PubMed] [Google Scholar]
  • 81. Moskowitz MA, Kiwak KJ, Hekimian K, et al. Synthesis of compounds with properties of leukotrienes C4 and D4 in gerbil brains after ischemia and reperfusion. Science 1984;224:886–889. [DOI] [PubMed] [Google Scholar]
  • 82. Minamisawa H, Terashi A, Katayama Y, et al. Brain eicosanoid levels in spontaneously hypertensive rats after ischemia with reperfusion: Leukotriene C4 as a possible cause of cerebral edema. Stroke 1988;19:372–377. [DOI] [PubMed] [Google Scholar]
  • 83. Mabe H, Nagai H, Suzuka T. Role of brain tissue leukotriene in brain oedema following cerebral ischaemia: Effect of a 5‐lipoxygenase inhibitor, AA‐861. Neurol Res 1990;12:165–168. [DOI] [PubMed] [Google Scholar]
  • 84. Aktan S, Aykut C, Ercan S. Leukotriene C4 and prostaglandin E2 activities in the serum and cerebrospinal fluid during acute cerebral ischemia. Prostaglandins Leukot Essent Fatty Acids 1991;43:247–249. [DOI] [PubMed] [Google Scholar]
  • 85. Bonventre JV, Huang Z, Taheri MR, et al. Reduced fertility and postischaemic brain injury in mice deficient in cytosolic phospholipase A2. Nature 1997;390:622–625. [DOI] [PubMed] [Google Scholar]
  • 86. Tabuchi S, Uozumi N, Ishii S, et al. Mice deficient in cytosolic phospholipase A2 are less susceptible to cerebral ischemia/reperfusion injury. Acta Neurochir Suppl 2003;86:169–172. [DOI] [PubMed] [Google Scholar]
  • 87. Ciceri P, Rabuffetti M, Monopoli A, et al. Production of leukotrienes in a model of focal cerebral ischaemia in the rat. Brit J Pharmacol 2001;133:1323–1329. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Shi WZ, Zhao CZ, Zhao B, et al. Aggravated inflammation and increased expression of cysteinyl leukotriene receptors in the brain after focal cerebral ischemia in AQP4‐deficient mice. Neurosci Bull 2012;28:680–692. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. Yu GL, Wei EQ, Wang ML, et al. Pranlukast, a cysteinyl leukotriene receptor‐1 antagonist, protects against chronic ischemic brain injury and inhibits the glial scar formation in mice. Brain Res 2005;1053:116–125. [DOI] [PubMed] [Google Scholar]
  • 90. Yu GL, Wei EQ, Zhang SH, et al. Montelukast, a cysteinyl leukotriene receptor‐1 antagonist, dose‐ and time‐dependently protects against focal cerebral ischemia in mice. Pharmacology 2005;73:31–40. [DOI] [PubMed] [Google Scholar]
  • 91. Saad MA, Abdelsalam RM, Kenawy SA, et al. Montelukast, a cysteinyl leukotriene receptor‐1 antagonist protects against hippocampal injury induced by transient global cerebral ischemia and reperfusion in rats. Neurochem Res 2015;40:139–150. [DOI] [PubMed] [Google Scholar]
  • 92. Shi QJ, Xiao L, Zhao B, et al. 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. [DOI] [PubMed] [Google Scholar]
  • 93. Zhao B, Zhao CZ, Zhang XY, et al. The new P2Y‐like receptor G protein‐coupled receptor 17 mediates acute neuronal injury and late microgliosis after focal cerebral ischemia in rats. Neuroscience 2012;202:42–57. [DOI] [PubMed] [Google Scholar]
  • 94. Tu XK, Yang WZ, Shi SS, et al. 5‐lipoxygenase inhibitor zileuton attenuates ischemic brain damage: Involvement of matrix metalloproteinase 9. Neurol Res 2009;31:848–852. [DOI] [PubMed] [Google Scholar]
  • 95. Silva BC, de Miranda AS, Rodrigues FG, et al. The 5‐lipoxygenase (5‐LOX) inhibitor Zileuton reduces inflammation and infarct size with improvement in neurological outcome following cerebral ischemia. Curr Neurovasc Res 2015;12:398–403. [DOI] [PubMed] [Google Scholar]
  • 96. Strom JO, Strid T, Hammarstrom S. Disruption of the alox5ap gene ameliorates focal ischemic stroke: Possible consequence of impaired leukotriene biosynthesis. BMC Neurosci 2012;13:146. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97. Wang G, Zhang J, Sun H, et al. Genetic variation in members of the leukotrienes biosynthesis pathway confers risk of ischemic stroke in Eastern Han Chinese. Prostaglandins Leukot Essent Fatty Acids 2012;87:169–175. [DOI] [PubMed] [Google Scholar]
  • 98. Ji R, Jia J, Ma X, et al. Genetic variants in the promoter region of the ALOX5AP gene and susceptibility of ischemic stroke. Cerebrovasc Dis 2011;32:261–268. [DOI] [PubMed] [Google Scholar]
  • 99. Sun H, Wu H, Zhang J, et al. A tagging SNP in ALOX5AP and risk of stroke: A haplotype‐based analysis among eastern Chinese Han population. Mol Biol Rep 2011;38:4731–4738. [DOI] [PubMed] [Google Scholar]
  • 100. Smith KJ, McDonald WI. The pathophysiology of multiple sclerosis: The mechanisms underlying the production of symptoms and the natural history of the disease. Philos Trans R Soc Lond B Biol Sci 1999;354:1649–1673. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101. Whitney LW, Ludwin SK, McFarland HF, et al. Microarray analysis of gene expression in multiple sclerosis and EAE identifies 5‐lipoxygenase as a component of inflammatory lesions. J Neuroimmunol 2001;121:40–48. [DOI] [PubMed] [Google Scholar]
  • 102. Arthur AT, Armati PJ, Bye C, et al. Genes implicated in multiple sclerosis pathogenesis from consilience of genotyping and expression profiles in relapse and remission. BMC Med Genet 2008;9:1–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103. Irkec C, Ercan S, Irkec M. Leukotrienes and neurological diseases. Mikrobiyol Bul 1989;23:342–347 (Article in Turkish). [PubMed] [Google Scholar]
  • 104. Neu I, Mallinger J, Wildfeuer A, et al. Leukotrienes in the cerebrospinal fluid of multiple sclerosis patients. Acta Neurol Scand 1992;86:586–587. [DOI] [PubMed] [Google Scholar]
  • 105. Yoshikawa K, Palumbo S, Toscano CD, et al. Inhibition of 5‐lipoxygenase activity in mice during cuprizone‐induced demyelination attenuates neuroinflammation, motor dysfunction and axonal damage. Prostaglandins Leukot Essent Fatty Acids 2011;85:43–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106. Marusic S, Leach MW, Pelker JW, et al. Cytosolic phospholipase A2 alpha‐deficient mice are resistant to experimental autoimmune encephalomyelitis. J Exp Med 2005;202:841–851. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107. Kalyvas A, Baskakis C, Magrioti V, et al. Differing roles for members of the phospholipase A2 superfamily in experimental autoimmune encephalomyelitis. Brain 2009;132:1221–1235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108. Thakker P, Marusic S, Stedman NL, et al. Cytosolic phospholipase A2alpha blockade abrogates disease during the tissue‐damage effector phase of experimental autoimmune encephalomyelitis by its action on APCs. J Immunol 2011;187:1986–1997. [DOI] [PubMed] [Google Scholar]
  • 109. Lee W, Su Kim H, Lee GR. Leukotrienes induce the migration of Th17 cells. Immunol Cell Biol 2015;93:472–479. [DOI] [PubMed] [Google Scholar]
  • 110. Wang L, Du C, Lv J, et al. Antiasthmatic drugs targeting the cysteinyl leukotriene receptor 1 alleviate central nervous system inflammatory cell infiltration and pathogenesis of experimental autoimmune encephalomyelitis. J Immunol 2011;187:2336–2345. [DOI] [PubMed] [Google Scholar]
  • 111. Kong W, Hooper KM, Ganea D. The natural dual cyclooxygenase and 5‐lipoxygenase inhibitor flavocoxid is protective in EAE through effects on Th1/Th17 differentiation and macrophage/microglia activation. Brain Behav Immun 2016;53:59–71. [DOI] [PubMed] [Google Scholar]
  • 112. Fumagalli M, Lecca D, Abbracchio MP. CNS remyelination as a novel reparative approach to neurodegenerative diseases: The roles of purinergic signaling and the P2Y‐like receptor GPR17. Neuropharmacology 2016;104:82–93. [DOI] [PubMed] [Google Scholar]
  • 113. Wang XY, Tang SS, Hu M, et al. Leukotriene D4 induces amyloid‐beta generation via CysLT(1)R‐mediated NF‐kappaB pathways in primary neurons. Neurochem Int 2013;62:340–347. [DOI] [PubMed] [Google Scholar]
  • 114. Tang SS, Wang XY, Hong H, et al. Leukotriene D4 induces cognitive impairment through enhancement of CysLT(1) R‐mediated amyloid‐beta generation in mice. Neuropharmacology 2013;65:182–192. [DOI] [PubMed] [Google Scholar]
  • 115. Herbst‐Robinson KJ, Liu L, James M, et al. Inflammatory Eicosanoids increase Amyloid precursor protein expression via activation of multiple neuronal receptors. Sci Rep 2015;5:18286. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116. Tang SS, Ji MJ, Chen L, et al. Protective effect of pranlukast on Abeta(1)(‐)(4)(2)‐induced cognitive deficits associated with downregulation of cysteinyl leukotriene receptor 1. Int J Neuropsychopharmacol 2014;17:581–592. [DOI] [PubMed] [Google Scholar]
  • 117. Lai J, Hu M, Wang H, et al. Montelukast targeting the cysteinyl leukotriene receptor 1 ameliorates Abeta1‐42‐induced memory impairment and neuroinflammatory and apoptotic responses in mice. Neuropharmacology 2014;79:707–714. [DOI] [PubMed] [Google Scholar]
  • 118. Qu T, Manev R, Manev H. 5‐Lipoxygenase (5‐LOX) promoter polymorphism in patients with early‐onset and late‐onset Alzheimer's disease. J Neuropsychiatry Clin Neurosci 2001;13:304–305. [DOI] [PubMed] [Google Scholar]
  • 119. Manev H, Manev R. 5‐Lipoxygenase (ALOX5) and FLAP (ALOX5AP) gene polymorphisms as factors in vascular pathology and Alzheimer's disease. Med Hypotheses 2006;66:501–503. [DOI] [PubMed] [Google Scholar]
  • 120. Sery O, Hlinecka L, Povova J, et al. Arachidonate 5‐lipoxygenase (ALOX5) gene polymorphism is associated with Alzheimer's disease and body mass index. J Neurol Sci 2016;362:27–32. [DOI] [PubMed] [Google Scholar]
  • 121. Listi F, Caruso C, Lio D, et al. Role of cyclooxygenase‐2 and 5‐lipoxygenase polymorphisms in Alzheimer's disease in a population from northern Italy: Implication for pharmacogenomics. J Alzheimers Dis 2010;19:551–557. [DOI] [PubMed] [Google Scholar]
  • 122. Di Francesco A, Arosio B, Gussago C, et al. Involvement of 5‐lipoxygenase in Alzheimer's disease: A role for DNA methylation. J Alzheimers Dis 2013;37:3–8. [DOI] [PubMed] [Google Scholar]
  • 123. Cipollone F, Mezzetti A, Fazia ML, et al. Association between 5‐lipoxygenase expression and plaque instability in humans. Arterioscler Throm Vasc Biol 2005;25:1665–1670. [DOI] [PubMed] [Google Scholar]
  • 124. Chu J, Pratico D. The 5‐lipoxygenase as a common pathway for pathological brain and vascular aging. Cardiovasc Psychiatry Neurol 2009;2009:174657. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125. Ikonomovic MD, Abrahamson EE, Uz T, et al. Increased 5‐lipoxygenase immunoreactivity in the hippocampus of patients with Alzheimer's disease. J Histochem Cytochem 2008;56:1065–1073. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126. Chu J, Pratico D. 5‐lipoxygenase as an endogenous modulator of amyloid beta formation in vivo. Ann Neurol 2011;69:34–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127. Puccio S, Chu J, Pratico D. Involvement of 5‐lipoxygenase in the corticosteroid‐dependent amyloid beta formation: In vitro and in vivo evidence. PLoS ONE 2011;6:e15163. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128. Chen YP, Zhang ZY, Li YP, et al. Syntheses and evaluation of novel isoliquiritigenin derivatives as potential dual inhibitors for amyloid‐beta aggregation and 5‐lipoxygenase. Eur J Med Chem 2013;66:22–31. [DOI] [PubMed] [Google Scholar]
  • 129. Chu J, Pratico D. Pharmacologic blockade of 5‐lipoxygenase improves the amyloidotic phenotype of an Alzheimer's disease transgenic mouse model involvement of gamma‐secretase. Am J Pathol 2011;178:1762–1769. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 130. Giannopoulos PF, Chu J, Joshi YB, et al. 5‐lipoxygenase activating protein reduction ameliorates cognitive deficit, synaptic dysfunction, and neuropathology in a mouse model of Alzheimer's disease. Biol Psychiatry 2013;74:348–356. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 131. Chu J, Lauretti E, Di Meco A, et al. FLAP pharmacological blockade modulates metabolism of endogenous tau in vivo. Transl Psychiatry 2013;3:e333. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132. Deng YM, Xie QM, Zhang SJ, et al. Changes of 5‐lipoxygenase pathway and proinflammatory mediators in cerebral cortex and lung tissue of sensitized rats. Acta Pharmacol Sin 2005;26:353–358. [DOI] [PubMed] [Google Scholar]
  • 133. Klegeris A, McGeer PL. Cyclooxygenase and 5‐lipoxygenase inhibitors protect against mononuclear phagocyte neurotoxicity. Neurobiol Aging 2002;23:787–794. [DOI] [PubMed] [Google Scholar]
  • 134. Kumar A, Sharma S, Prashar A, et al. Effect of licofelone–a dual COX/5‐LOX inhibitor in intracerebroventricular streptozotocin‐induced behavioral and biochemical abnormalities in rats. J Mol Neurosci 2015;55:749–759. [DOI] [PubMed] [Google Scholar]
  • 135. Manev H, Uz T, Qu T. Early upregulation of hippocampal 5‐lipoxygenase following systemic administration of kainate to rats. Restor Neurol Neurosci 1998;12:81–85. [PubMed] [Google Scholar]
  • 136. Kumar A, Prakash A, Pahwa D, et al. Montelukast potentiates the protective effect of rofecoxib against kainic acid‐induced cognitive dysfunction in rats. Pharmacol Biochem Behav 2012;103:43–52. [DOI] [PubMed] [Google Scholar]
  • 137. Chou VP, Holman TR, Manning‐Bog AB. Differential contribution of lipoxygenase isozymes to nigrostriatal vulnerability. Neuroscience 2013;228:73–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138. Kang KH, Liou HH, Hour MJ, et al. Protection of dopaminergic neurons by 5‐lipoxygenase inhibitor. Neuropharmacology 2013;73:380–387. [DOI] [PubMed] [Google Scholar]
  • 139. Wang H, Shi Q, Shi W, et al. Expression and distribution of cysteinyl leukotriene receptors CysLT1R and CysLT2R, and GPR17 in brain of Parkinson disease model mice. Zhejiang Da Xue Xue Bao Yi Xue Ban 2013;42:52–60 (Article in Chinese). [DOI] [PubMed] [Google Scholar]
  • 140. Chen L, Yang Y, Li CT, et al. CysLT2 receptor mediates lipopolysaccharide‐induced microglial inflammation and consequent neurotoxicity in vitro. Brain Res 2015;1624:433–445. [DOI] [PubMed] [Google Scholar]
  • 141. Lenz QF, Arroyo DS, Temp FR, et al. Cysteinyl leukotriene receptor (CysLT) antagonists decrease pentylenetetrazol‐induced seizures and blood–brain barrier dysfunction. Neuroscience 2014;277:859–871. [DOI] [PubMed] [Google Scholar]
  • 142. Rehni AK, Singh TG. Modulation of leukotriene D4 attenuates the development of seizures in mice. Prostaglandins Leukot Essent Fatty Acids 2011;85:97–106. [DOI] [PubMed] [Google Scholar]
  • 143. Fleck J, Temp FR, Marafiga JR, et al. Montelukast reduces seizures in pentylenetetrazol‐kindled mice. Braz J Med Biol Res 2016;49:e5031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144. Takahashi Y, Imai K, Ikeda H, et al. Open study of pranlukast add‐on therapy in intractable partial epilepsy. Brain Dev 2013;35:236–244. [DOI] [PubMed] [Google Scholar]
  • 145. Bertin J, Jalaguier P, Barat C, et al. 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;454–455:128–138. [DOI] [PubMed] [Google Scholar]
  • 146. Zhu L, Maruvada R, Sapirstein A, et al. Arachidonic acid metabolism regulates Escherichia coli penetration of the blood‐brain barrier. Infect Immun 2010;78:4302–4310. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147. Di Guardo G, Falconi A, Di Francesco A, et al. Western blot expression of 5‐lipoxygenase in the brain from striped dolphins (stenella coeruleoalba) and bottlenose dolphins (tursiops truncatus) with or without encephalitis/meningo‐encephalitis of infectious nature. J Biol Regul Homeost Agents 2015;29:245–250. [PubMed] [Google Scholar]
  • 148. Kalonia H, Kumar P, Kumar A, et al. Protective effect of montelukast against quinolinic acid/malonic acid induced neurotoxicity: Possible behavioral, biochemical, mitochondrial and tumor necrosis factor‐alpha level alterations in rats. Neuroscience 2010;171:284–299. [DOI] [PubMed] [Google Scholar]
  • 149. Yu XB, Dong RR, Wang H, et al. Knockdown of hippocampal cysteinyl leukotriene receptor 1 prevents depressive behavior and neuroinflammation induced by chronic mild stress in mice. Psychopharmacology 2016;233:1739–1749. [DOI] [PubMed] [Google Scholar]

Articles from CNS Neuroscience & Therapeutics are provided here courtesy of Wiley

RESOURCES