Next Article in Journal
A Peptide Inhibitor of the Human Cytomegalovirus Core Nuclear Egress Complex
Previous Article in Journal
An Evaluation of the Drug Permeability Properties of Human Cadaveric In Situ Tympanic and Round Window Membranes
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Leukotrienes vs. Montelukast—Activity, Metabolism, and Toxicity Hints for Repurposing

by
Cátia F. Marques
1,
Maria Matilde Marques
1,2 and
Gonçalo C. Justino
1,*
1
Centro de Química Estrutural, Institute of Molecular Sciences, Instituto Superior Técnico, Universidade de Lisboa, 1049-001 Lisboa, Portugal
2
Departamento de Engenharia Química, Instituto Superior Técnico, Universidade de Lisboa, 1049-001 Lisboa, Portugal
*
Author to whom correspondence should be addressed.
Pharmaceuticals 2022, 15(9), 1039; https://doi.org/10.3390/ph15091039
Submission received: 22 July 2022 / Revised: 17 August 2022 / Accepted: 19 August 2022 / Published: 23 August 2022
(This article belongs to the Section Medicinal Chemistry)

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.

Graphical Abstract

1. Introduction

Montelukast (MTK) is an antagonist of the cysteinyl leukotrienes receptor 1 and is routinely used in the management of asthma symptoms among adults and children. Its systemic anti-inflammatory actions, which are particularly important in the brain tissues, are at the onset of various clinical studies focused on the repurposing of this drug for various other diseases, aimed particularly at Alzheimer’s and Parkinson’s diseases. However, this repurposing clashes with neuropsychiatric adverse drug reactions elicited by the drug. Starting with a brief overview of the biochemistry of leukotrienes, this work reviews the current knowledge on montelukast.

2. Cysteinyl Leukotrienes—Multifunctional Inflammation Mediators

2.1. Cysteinyl Leukotrienes and Their Receptors

Initially described as the slow-reacting substances of anaphylaxis, leukotrienes (LTs) are pro-inflammatory lipid mediators derived from arachidonic acid [1,2]. These mediators are synthesized mainly in cells from the innate immune system (e.g., polymorphonuclear leukocytes, macrophages, mast cells, and brain microglia) following activation by immune and non-immune stimuli such as infection, tissue injury, allergens, and exercise (Figure 1). Upon cell activation, the cytosolic calcium concentration increases, and the cytosolic phospholipase A2 (cPLA2) and 5-lipoxygenase (5-LOX) enzymes are activated and translocated to the nuclear envelope. There, cPLA2 cleaves glycerophospholipids, releasing arachidonic acid (AA), which is converted to the acyclic hydroperoxide 5(S)-hydroperoxyeicosatetraenoic acid (5-HpETE) by 5-LOX-mediated oxidation upon LOX activation by 5-LOX activating protein (FLAP); 5-HpETE, in turn, undergoes dehydration to the unstable conjugated triene epoxide leukotriene A4 (LTA4), the first metabolite in the leukotriene pathway. LTA4 is a short-lived intermediate that can undergo conjugate addition of water to form leukotriene B4 (LTB4) or conjugation with glutathione by LTC4 synthase to form leukotriene C4 (LTC4, an S-glutathionyl LT). LTB4 and LTC4 are transported to the extracellular space mainly by multidrug resistance proteins, namely through MRP4 (LTB4) and MRP1 (LTC4) [3,4], where cleavage of LTC4 to leukotriene D4 (LTD4) and subsequently to leukotriene E4 (LTE4) takes place. LTD4, an S-cysteinyl LT, is synthesised from LTC4 by a γ-glutamyl transpeptidase (GGT)-mediated cleavage, whereas LTE4 results from the cleavage of LTD4 by a membrane-bound dipeptidase [5,6,7,8,9,10,11,12,13,14,15,16,17,18].
Figure 1 summarizes the biosynthesis of leukotrienes as well as their interactions with the leukotriene receptors.
LTB4 is a pro-inflammatory LT that acts on human polymorphonuclear leukocytes (PMNLs) such as neutrophils, via G protein-coupled receptors B-LT1 or B-LT2, triggering chemotaxis and the subsequent activation of the inflammatory response. LTC4, LTD4, and LTE4 constitute a group of cysteinyl leukotrienes (CysLTs) that act through G protein-coupled cell surface receptors, of which the two classical receptors are the cysteinyl leukotriene receptors 1 (CysLTR1) and 2 (CysLTR2). LTC4 is an agonist of CysLTR1 whereas LTD4 binds CysLTR1 and CysLTR2. LTE4 is described as an agonist of CysLTR3 (also known as GPR99 receptor) and of the purinergic receptors GPR17 and P2Y12 [5,6,7,8,9,10,11,12,13,14,15,16,17,18].
Cysteinyl leukotriene receptors (CysLTRs) are involved in the pathophysiology of various respiratory allergic diseases, including bronchial asthma, exercise- and aspirin-induced asthma, and allergic rhinitis, as well as atopic dermatitis, allergic conjunctivitis, and anaphylaxis, exhibiting a large overlap with the B-LT receptors, but allowing a finely tuned immune response [11,12,13,20,21]. Receptor engagement by CysLTs promotes bronchoconstriction, vascular leakage, and neutrophil extravasation to inflammation sites [7]. CysLTR1 is expressed in most human tissues, particularly in the appendix, oesophagus, gall bladder, lung, lymph nodes, spleen, and urinary bladder. The affinity of leukotrienes to this receptor varies in the order LTD4 > LTC4 > LTE4. This receptor is sensitive to classical antagonists (Figure 2) such as montelukast (MTK, Singulair®), zafirlukast (Accolate®), pranlukast (Onon®, Azlaire®), pobilukast, and MK571, all members of the Lukast group (cysteinyl leukotriene receptor antagonists).
CysLTR2 is predominantly expressed in the spleen, heart, brain, and adrenal gland, and its affinity strength is LTC4 = LTD4 > LTE4. HAMI3379 (Figure 2) was identified as a potent and selective CysLTR2 receptor antagonist [22]. To our knowledge, only two dual inhibitors of both CysTR1 and CysLTR2 are reported—BAY-u9773 and gemilukast (Figure 2). However, BAY-u9773 is neither very potent nor selective for human CysLTs [11,12,20,21,23] and gemilukast did not show outcome differences when compared with MTK [24,25]. Figure 2 also shows the experimental IC50 values available for these compounds.
Figure 2. Cysteinyl leukotrienes receptor antagonists. CysLTR1 antagonists (montelukast, zafirlukast, pranlukast, pobilukast, and MK571), CysLTR2 antagonists (HAMI3379) and dual antagonists (BAY-u9773 and gemilukast) are shown. Experimental IC50 values available from the literature are also given in the inset table [22,25,26,27,28,29,30].
Figure 2. Cysteinyl leukotrienes receptor antagonists. CysLTR1 antagonists (montelukast, zafirlukast, pranlukast, pobilukast, and MK571), CysLTR2 antagonists (HAMI3379) and dual antagonists (BAY-u9773 and gemilukast) are shown. Experimental IC50 values available from the literature are also given in the inset table [22,25,26,27,28,29,30].
Pharmaceuticals 15 01039 g002
Besides these classical receptors, three other receptors are associated with the leukotriene cascade—GPR99, P2Y12, and GPR17.
GPR99, or OXGR1, is an α-ketoglutarate receptor that was originally thought to be a P2Y receptor [31]. This receptor is expressed in the kidney, placenta, trachea, salivary glands, lungs, and smooth muscle cells, as well as in some brain regions; in addition to its effects on acid–base homeostasis, it is also involved in axon growth [32,33,34,35]. GPR99 is considered the third CysLT receptor (CysLTR3) due to its high affinity for LTE4. No antagonists are currently available for this receptor [32,36].
P2Y12 is an adenosine diphosphate receptor that also mediates LTE4-dependent pulmonary inflammation (but not the LTD4 response) [37]. This receptor is mainly expressed in platelets and microglia, where it triggers platelet activation and blood clotting, and induces microglial chemotaxis in situations of central nervous system (CNS) injury [38,39,40,41]. P2Y12 is also associated with some asthma symptoms, namely with eosinophilic inflammation and airway hyper-responsiveness [42,43]. The P2Y12 receptor is blocked by anti-platelet drugs such as clopidogrel, prasugrel, and ticagrelor [44].
Lastly, GPR17 is a uracil nucleotide P2Y receptor expressed in the brain that also binds CysLTs [12,14,45,46,47,48,49,50]. This receptor is described as a sensor of neuronal damage, being activated by nucleotides and CysLTs released in the damaged area and plays a dual role depending on its surroundings: under physiological conditions, GPR17 contributes to the differentiation and maturation of oligodendrocytes, whereas under pathological conditions it mediates demyelination and apoptosis [51,52,53,54,55,56]. GPR17 is described as a putative negative regulator of CysLTR1 [57]. The CysLTR1 inhibitors pranlukast and montelukast are also antagonists of this receptor [46,58,59].

2.2. Leukotrienes in the Brain

The potential of leukotrienes as pro-inflammatory lipid mediators, described above, together with the pattern of expression of their receptors in different organs, has led to the suggestion that LTs play an important role in the central nervous system. In fact, recent advances have associated inflammation with some brain pathologies such as multiple sclerosis, Alzheimer’s disease, Parkinson’s disease, brain ischemia, and epilepsy, among others, and leukotrienes are thought to play a role in this process [60,61].
Despite having been originally found in leukocytes, leukotrienes are also present in the brain. Not only is the 5-LOX enzyme widely distributed in various brain regions (e.g., cortex, hippocampus, and cerebellum), but CysLTs are also produced by vascular endothelial cells, neurons, and glial cells upon LTA4 expression by activated neutrophils [47]. CysLTR1 is widely expressed in the cortex, hippocampus, and nigrostriatum, as well as in cerebrovascular endothelial cells, astrocytes, microglia, and several types of neurons. On the other hand, CysLTR2 is expressed in the cortex, hippocampus, substantia nigra, astrocytes, microglia, and neurons [62,63,64,65,66]. These receptors are usually weakly expressed unless activated by pathological stimuli [66]. Some studies have shown that the exposure of neurons to acute neuronal injury is associated with upregulated levels of CysLTR1 and CysLTR2, and with increased blood–brain barrier (BBB) permeability. Once activated, CysLT receptors will trigger an inflammatory cascade, activating pro-inflammatory cytokines and inflammation, ultimately leading to neuronal damage [62,64,65,67].

2.2.1. Leukotrienes: Role in Neuroinflammation

Neuroinflammation is a complex biological response of the brain and spinal cord mediated by the production of pro-inflammatory cytokines (IL-1β, IL-6, and TNF-α), chemokines (CCL2, CCL5, and CXCL1), reactive oxygen species (ROS), and other mediators (NO, prostaglandins, and leukotrienes) [68,69,70]. This biological response is associated with restoration of homeostatic balance, in order to eliminate and repair the initial cause of cell injury, and can be classified as acute (seconds to days) or chronic [68,71].
An acute inflammatory response is an adaptive response, usually beneficial, meant to protect tissues from a specific injury as trauma or infection [71]. As presented in Figure 3, in situations of acute inflammation, the immune system priorities are neuroprotection, tissue repair, and neuroplasticity. When the brain is exposed to immune signals after any infection, microglia and astrocytes are activated and neuroinflammatory cytokines such as IL-1β, TNF-α, and IL-6 are expressed to sustain the inflammatory response. This response is short and transient, and no severe effects take place [68]. Brain development and plasticity are other positive aspects of neuroinflammation. Neurons, astrocytes, and glia cells are involved in neurotransmission through the modulatory effect of cytokines and neuromodulators such as IL-1β, IL-6, TNF-α, NF-κB, and glutamate [68,72]. Brain tissue repair can also be stimulated through the activation of macrophages, lymphocytes, and microglia, which promotes angiogenesis, axon regeneration, myelin clearance, and oligodendrocyte regeneration [68,73,74,75]. Lastly, immune system training through immune pre-conditioning or euflammation allows modulation of the microglia response against hyper-inflammatory conditions, protecting the brain from CNS injuries [68,76,77].
However, if the acute inflammation response fails and the inflammation process persists, chronic inflammation ensues with a long-lasting maladaptive or defective response that could destroy tissues and compromise the immune response (Figure 3) [70,71]. Characteristically, there is an increased production of cytokines (IL-1 and TNF-α), reactive oxygen species (ROS), and other inflammatory mediators (e.g., inducible nitric oxide synthase, iNOS), associated with the activation of microglia cells, and consequent expression of more pro-inflammatory cytokines and chemokines in the brain [68]. This activation could be caused by noradrenergic signalling, inflammasome activation, and ATP release [78,79,80]. Microglia activation is also involved in the recruitment of monocytes from the bone marrow to the brain and is linked to anxiety-like behaviour and to the development of mood disorders [68,81].
The normal ageing process is one example of the disruption of the communication pathways between the brain and the immune system, leading to chronic neuroinflammation. During ageing, there is an increase in inflammatory (e.g., IL-1β and IL-6) and a decrease in anti-inflammatory (e.g., IL-10 and IL-4) cytokines that results in damage to the nervous system and the onset of neurodegenerative diseases [68].
It has been shown that the leukotriene receptors CysLTR1 and CysLTR2 in different brain cells, namely microglia (known as the brain’s immune system), astrocytes, and several types of neurons, are upregulated in response to brain injury such as brain ischemia, Alzheimer’s disease, and Parkinson’s disease [62,63,64,65,82,83,84,85,86,87,88]. The modulation of these receptors is associated not only with the outcome of acute inflammation but also with the restoring of homeostasis during chronic inflammation [62,63,64,65,82,83,84,85,86,87,88].
Although the mechanisms of action are still poorly understood, evidence supports the relationship between leukotrienes and neuroinflammation, suggesting the use of leukotriene antagonists as a possible therapeutic strategy in neuroinflammation, given that antagonists of either CysLTR1 or CysLTR2 display wide multi-target anti-inflammatory activity [66]. Both receptors are expressed at low levels in multiple brain regions, but are upregulated following injury, as observed in various experimental models of ischemia and Alzheimer’s and Parkinson’s diseases [65,82,83,84,89]. Interestingly, silencing the expression of the genes coding for these two receptors leads to in vivo protection against lipopolysaccharide- and ischemia-induced brain inflammation and injury [87,88]. Although this strategy needs to be further explored, it could be a very promising therapeutic approach to the improvement of symptoms (or even disease treatment) in patients who suffer from neurodegenerative disorders and have no alternative therapy to manage the debilitating symptoms characteristic of neurodegeneration.

2.2.2. Leukotrienes in Neuro-Signalling Pathways

Message transmission between neurons results from an electrical impulse (action potential) that causes the release of neurotransmitters into the synaptic cleft. After crossing the synaptic cleft, neurotransmitters will reach their receptors on the postsynaptic side to excite or inhibit the target neuron. Excitatory synaptic transmission is mainly assured by L-glutamate, whereas γ-aminobutyric acid (GABA) is the major neurotransmitter involved in the inhibitory synaptic response. In addition to these neurotransmitters, there are other molecules involved in signalling and neuromodulation, such as acetylcholine, monoamines (e.g., dopamine, adrenaline, serotonin, and histamine), purines (e.g., adenosine), and neuropeptides [89].
A close relationship between neuroinflammation and neuro-signalling pathways has been proposed. One example is the involvement of excitotoxicity in neuroinflammation: an exacerbated or prolonged activation of glutamate receptors, particularly the N-methyl-D-aspartic acid receptors (NMDA), causes an increase in calcium influx into the neurons. This increase of intracellular calcium levels leads to a neurotoxic response, including the activation of the AA pathway, that can lead to the loss of neuronal function and, ultimately, cell death [90]. Studies involving CysLTR antagonists showed that pranlukast was able to inhibit NMDA-induced CysLTR1 expression, leading to a decrease in excitotoxic cell death [91]. Montelukast also presented a strong anti-excitotoxicity effect, as well as anti-inflammatory and neuroprotective properties [83].
Dopamine reuptake is also associated with the leukotriene pathway. Inhibition of the 5-LOX activating protein (FLAP) is associated with the improved integrity of dopaminergic neurons [92].

2.2.3. The Leukotriene Link between Stress and Depression

As suggested in Figure 3, depression can result from chronic neuroinflammation. Not only pro-inflammatory cytokines (e.g., IL-1β and TNF-α) were found to be dysregulated in depression patients, but also IL-1β, IL-6, TNF-α, or lipopolysaccharide (LPS) administration in animal models led to depression- and anxiety-like behaviours [93,94,95,96].
Stress stimuli led to an increase in calcium concentration, releasing AA after cPLA2 activation by phosphorylation [97]. Once released, AA is used to synthesise leukotrienes (Figure 1) and prostaglandins. A study using mice in which the cysltr1 gene was silenced in the hippocampus suggested that the absence of CysLTR1 prevents the development of neuroinflammation and of a depressive-like phenotype [98]. The effects observed upon blocking the same receptors in a mouse lipopolysaccharide-induced neuroinflammation model support those previous results [99]. Inhibition of the 5-LOX enzyme has also been associated with a relief of depression-like behaviour [100].

2.2.4. The Role of Leukotrienes in Neurodegenerative Diseases

Besides their role in inflammation, leukotrienes are also involved in some of the most characteristic hallmarks of neurodegenerative disorders (Figure 4): neuronal cell death, neuroinflammation, altered neurogenesis, and disrupted blood–brain barrier and vascular system, among others.
The clear association between neuroinflammation and Alzheimer’s and/or Parkinson’s disease led to the study of the role of CysLTs pathways and receptors in these diseases.
Alzheimer’s disease (AD, described in more detail in Section 4.1) is a neurodegenerative disease characterized by memory loss and dementia. There is evidence for CysLTR1 involvement in AD, leading to amyloidogenesis and neuroinflammation. In particular:
(1) In an AD mouse model (APP/PS1 double transgenic, overexpressing mutated forms of human amyloid precursor protein, APP, and presenilin 1), the expression of CysLTR1 was found to increase with ageing, and to correlate with Aβ deposits and behaviour deficits [84,101];
(2) LTD4 upregulates APP, β-, and γ-secretase levels, and facilitates Aβ amyloid accumulation via the CysLTR1-mediated NF-κB pathway [102,103,104].
Aggregated Aβ1–42 is known to cause AD-like neurotoxicity and cognitive deficiency, associated with pro-inflammatory cytokine production (TNF-α, IL-1β) and increased cell apoptosis [84,105,106]. Additional studies also revealed that Aβ plaques are associated with an increased oxidative stress status. Oxidative stress is known to upregulate cPLA2 activity, leading to an increased release of arachidonic acid metabolites [66]. These responses are inhibited by Lukast drugs (pranlukast, montelukast, and zafirlukast), suggesting that CysLTR1 is a pro-inflammatory regulator and is involved in AD initiation and progression [66,84,105,106].
Parkinson’s disease (PD, described in more detail in Section 4.2) is also a neurodegenerative disorder characterised by the progressive degeneration and loss of dopaminergic neurons. Inflammation induction in PD models (with rotenone or lipopolysaccharide) leads to microglia activation, increasing the production of the pro-inflammatory cytokines TNF-α, IL-1β, and IL-6, and brain inflammation, leading to dopaminergic neuronal loss [47,66,107,108,109,110]. This action was inhibited by montelukast via the CysLTR1-mediated p38 MAPK/NF-κB pathway [82,107,111], and also by selective inhibition or knockout of CysLTR2 [86], suggesting that CysLTR1 and CysLTR2 could be strategic targets against PD. CysLTR1, as well as 5-LOX, are found to be upregulated in mouse PD models [92], further strengthening the hypothesis that the LT pathway contributes to the progression of PD.
In conclusion, leukotrienes play an important role in the progression of neurodegenerative disorders. Receptors involved in the different steps of the LT cascade interfere with the inflammatory process, which is partially responsible for the development of the characteristic hallmarks of AD and PD. For this reason, targeting the CysLT pathway seems to be a promising strategy to delay the progression of these disorders.

3. A Cysteine Leukotriene Receptor Antagonist Known as Montelukast

The World Health Organization (WHO, Geneva, Switzerland) estimated that in 2019 more than 262 million people suffered from asthma, a pulmonary disorder that causes lung inflammation and tightening of the muscles around small airways [112]. The number of people suffering from this disorder is expected to increase, since a wide range of environmental risks is associated with asthma development, including tobacco smoking, pollution, and environmental allergens and irritants [112]. With no current treatment available, montelukast is broadly used in symptom management in adults and children.
Montelukast (MTK, 1-([(1(R)-(3-(2-(7-chloro-2-quinolinyl)-(E)-ethenyl)phenyl)-3-(2-(1-hydroxy-1-methylethyl)phenyl)propyl)thio]methyl) cyclopropylacetic acid, Figure 5), widely used in asthma management and allergic rhinitis, is a potent antagonist of CysLTR1, a receptor with high affinity for the leukotriene, LTD4. As indicated above, CysLTRs modulate the synthesis of the leukotrienes from arachidonic acid. They are involved in the pathology of various allergic diseases of the respiratory system, including bronchial asthma, exercise- and aspirin-induced asthma, and allergic rhinitis. These lead to airway constriction, smooth muscle contraction, and alterations in the inflammatory processes, such as neutrophil extravasation to the site of inflammation [113,114,115,116].
In addition to CysLTR1, targeted in asthma management, recent studies have identified further MTK targets that could be exploited against other pathologies, particularly in the central nervous system. MTK has been identified as an inhibitor of 5-LOX [117], and as an antagonist of the CysLTR2, P2Y12 [118], and GPR17 [47] receptors.

3.1. Montelukast Metabolism and Bioavailability

The sodium salt of MTK has been available since 1995 as Singulair® (Merck Sharp & Dohme, Kenilworth, NJ, USA) and has been increasingly prescribed in recent years [119]. However, MTK metabolism is still poorly understood.
The first metabolic studies of this drug were performed on healthy volunteers treated with 14C-MTK. Samples from blood, urine, faeces, bile, and gastric juices were collected during the clinical trial and eight MTK metabolites were identified (Figure 5) an acyl glucuronide (M1), a sulfoxide (M2), a phenol (M3), a dicarboxylic acid (M4), and hydroxylated metabolites at positions 21 (M5a/b, where a and b correspond to the 21-S and -R configurations) and 36 (M6a/b, with no assignment of the specific C36 configuration) [120]. According to the authors of the study, MTK is mainly excreted in the faeces (86% of the administered dose) and only 0.2% is excreted via urine. All identified metabolites were found in bile samples, where M5a was more abundant than M5b. M5 and M6 were also identified in plasma samples, with M6a being more abundant than M6b [120].
Clinical trial data were complemented with early in vitro studies, which showed that cytochrome P450 (CYP) enzymes are responsible for the phase I MTK metabolism, whereas flavin-containing monooxygenases (FMO) have little or no activity on this substrate [121,122].
Additional metabolic and inhibition assays have been described to identify further metabolites formed during MTK metabolism (phase I and II), as well as the active isoforms. Human liver microsomes, recombinant CYP enzymes (CYP1A2, 2A6, 2B6, 2C8, 2C9, 2C19, 2D6, 2E1, 3A4, and 3A5), UGT enzymes (UGT1A1, 1A3, 1A4, 1A6, 1A7, 1A8, 1A9, 1A10, 2B4, 2B7, 2B10, 2B15, and 2B17), and isoform-specific inhibitors were tested [120,121,122,123,124]. Only a new MTK ether glucuronide metabolite (M-glucuronide, Figure 5) was found in those studies.
The major enzymes responsible for metabolite production are identified in Figure 5. Table 1 shows the relative contribution of each CYP isoform toward the final products. CYP2C8 is mentioned to be the most relevant CYP involved in MTK metabolism, responsible for 70% of MTK oxidative metabolic clearance [123].
Figure 5. Human MTK metabolic pathways. The presented metabolites were identified in bile (M1, M-glucuronide, M2, M3, M4, and M6) and in plasma (M5 and M6) from healthy volunteers [120,121,122,123,125]. Atom numbering used in this work is included, in agreement with the literature numbering system used for montelukast.
Figure 5. Human MTK metabolic pathways. The presented metabolites were identified in bile (M1, M-glucuronide, M2, M3, M4, and M6) and in plasma (M5 and M6) from healthy volunteers [120,121,122,123,125]. Atom numbering used in this work is included, in agreement with the literature numbering system used for montelukast.
Pharmaceuticals 15 01039 g005
Briefly, MTK sulfoxide (M2) is produced mainly by CYP3A4 whereas M3 (25-OH-MTK) is obtained by the action of the CYP2C8, CYP2C9, CYP3A4, and CYP2C19 isoforms [121,122,123,124]. M5 (21-OH-MTK) results from CYP3A4 and CYP3A5 metabolism [121,122,123,124]. The dihydroxylated metabolite (M6, 36-OH-MTK), precursor to the dicarboxylate (M4), is produced by CYP2C8 and CYP2C9 [121,122,123,124], whereas M4 itself results from CYP2C8 catalysis [123]. M4 was considered to be the major metabolite by Balani et al. [120], whereas Cardoso et al. [124] consider M6 as the most abundant. For VandenBrink et al. [122], M2, M5, and M6 are the major ones. However, M2 is not a consensual metabolite since it is also an MTK impurity [123,124].
Only two phase II MTK metabolites have so far been identified by mass spectrometry: the M1 glucuronide (major) and the M-glucuronide (minor), both stemming from glucuronidation of the parent drug. UGT1A3 is reported to be the most active UDP-glucuronosyltransferase isoform involved in MTK glucuronidation [124,125].
MTK is characterized by a 60–70% bioavailability and high plasma protein binding capacity (>99%) and displays little or no gender effect on its pharmacokinetic properties [126,127]. No metabolism differences are reported between adults and children [121].
Despite the unfavourable absorption properties, such as high lipophilicity and high plasma protein binding, the efficiency of MTK transport across membranes, including the blood–brain barrier, remains unknown. Even though some researchers have identified MTK as a substrate of the organic anion transporting polypeptide 2B1 (OATP2B1) transporter, which is expressed in the blood–brain barrier, recent studies failed to confirm this observation [128,129,130,131].

3.2. Adverse Drug Reactions Related to Montelukast Administration

Montelukast belongs to the Lukast drug family, whose members are considered safe and well-tolerated drugs, suitable for long-term administration, with low toxicity and relatively low adverse side effects [116]. However, during post-marketing surveillance, some reports of adverse effects caused by monotherapy and co-adjuvant therapy with MTK emerged, motivating the US FDA to require a boxed warning regarding montelukast use and the occurrence of neuropsychiatric events. Since March 2020, the FDA recommends the use of alternative drugs, restricting MTK to patients with an inadequate response or intolerance to other therapies [132,133,134].
According to the WHO global database for adverse drug reactions (ADRs), Vigibase [135], 26,253 reports were filed until August 2021, and 22% of the reported ADRs occurred in children between the ages of 2 and 11. Psychiatric and nervous system disorders are the most reported, along with hepatobiliary, pancreatic, and uropoietic disorders, and immune system dysregulation. Although the number of reported ADRs is considerable, no underlying mechanisms have been proposed.
It is important to highlight the limitations of the ADR reporting system. Since ADR reporting is voluntary and patients and doctors only report when a correlation between a drug and ADRs is suspected, it is expected that MTK-related ADRs are underreported. On the other hand, some of the neuropsychiatric events experienced by patients are not exclusively correlated with MTK, but also with other physiopathologic, economic, and social conditions such as, for example, depression and sleep deprivation.
MTK toxic events are described in more detail in the following sections.

3.2.1. Neuropsychiatric and Nervous System Disorders

A growing number of MTK ADRs has been reported in the literature, focusing on neuropsychiatric aspects, especially anxiety and sleep disorders [136,137,138,139,140,141,142].
In 2009, a total of 48 reports of psychiatric disorders in children were found in the Swedish ADR database SWEDIS. Nightmares, general anxiety, aggressiveness, sleep disorders, insomnia, irritability, hallucination, hyperactivity, and personality disorder were some of the most reported ADRs. Approximately 50% of these effects occurred in children under 3 years old and, in 80% of the reports, ADRs developed within 1 week from the first MTK administration [143]. Later, a cohort of 14,670 individual case safety reports, of which 2630 corresponded to children and adolescents younger than 18 years old, were reviewed in 2015. The main conclusions highlighted children as the most likely to experience montelukast ADRs: sleep disorders were mostly reported in children younger than 2 years old; depression and anxiety signs in children between 2 and 11 years; and suicidal behaviour and depression/anxiety in adolescents between 12 and 17 years. Surprisingly, achieved suicides were more reported in children than adolescents or adults [137]. Between 2012 and 2017, an observational study in a Spanish paediatric hospital concluded that 5.7% of children under 15 years old experienced ADRs, mainly insomnia, hyperactivity, and nightmares, which disappeared after MTK discontinuation [144].
Isolated cases of well-defined neuropsychiatric events in children and adults taking montelukast are also described in the literature. A 9-year-old boy experienced sleepwalking, sleep disturbance, bruxism, and anxiety during MTK treatment. After MTK withdrawal, the symptoms resolved without further intervention [139]. Another case described a 13-year-old who experienced hallucinations that stopped 48 h after MTK withdrawal [138]. A 16-year-old girl who was medicated with MTK reported parasomnias (sleeptalking and sleepwalking) during two attempts at MTK treatment. Symptoms stopped after MTK withdrawal for both attempts [145]. A 29-year-old asthmatic woman suffered from visual and auditory hallucinations, which stopped two days after MTK withdrawal [146]. An HIV-positive female patient reported neuropsychiatric disturbance including sleep disorders, vivid dreams, irritability, and confusion after adding MTK to her usual medication (efavirenz) [147]. In this case, doctors suspected drug–drug interaction and drug competition between CYP isoforms involved in MTK metabolism [147].
The association between MTK and ADRs is in permanent evaluation. A recent study reports that the risk for psychiatric adverse events is greater in patients with past psychiatric history, and no association between depression or self-harm events and hospitalizations was identified [134]. Regarding MTK and suicide ideation, it remains a non-consensual subject among the scientific community, with some studies establishing a relationship between them and others denying it [148,149,150]. There is even a study suggesting that MTK may reduce the risk of suicide [151].
Summing up, both children and adults seem to develop psychiatric adverse side effects during montelukast treatment, especially children. Usually, the symptoms tend to disappear after drug withdrawal. However, it is important to understand the mechanisms underlying these ADRs in order to improve treatment and risk–benefit assessment, and to prevent dangerous outcomes.

3.2.2. Hepatobiliary, Pancreatic, and Uropoietic Disorders

Contrary to the neuropsychiatric ADRs, the hepatotoxicity of montelukast occurs mainly in adults. Usually, patients are polymedicated and the relationship between MTK and ADRs was established based on time exposure and drug exposure and withdrawal.
Patients between 22 and 76 years old medicated with MTK developed acute pancreatitis, hypercholesterolemia, and hypertriglyceridemia [152], haematuria [153], and hepatomegaly [154]. Vomiting, icterus, and high levels of liver biomarkers (aminotransferase, bilirubin, and alkaline phosphatase) were also associated with MTK treatment, with an underlying related immune-mediated mechanism of liver injury [155]. All patients improved their condition after MTK withdrawal.
Regarding children, there are clinical cases describing hepatitis, nausea, vomiting, abdominal pain, and high levels of liver biomarkers [156], as well as hepatocellular injury [157]. Children also recovered after MTK withdrawal.
With these examples in mind, kidney and renal function should be monitored in patients on montelukast therapy. The risk of hypertriglyceridemia can be harmful not only for cardiovascular risk patients but also for healthy patients [133].

3.2.3. Skin and Subcutaneous Tissue Disorders

Angioedema and urticaria, conditions with a strong inflammatory component, are the most commonly reported skin disorders in patients medicated with MTK [158,159]. A case of a child with erythematous and bullous eruption in the lower extremities after MTK treatment was also reported [160]. In all cases, symptoms promptly resolved upon stopping the treatment with MTK; in one case, symptom reappearance upon MTK re-introduction clearly established a link between MTK and the observed side effects [159].

3.2.4. Immune System Disorders

Immune system disorders associated with montelukast therapy are rare and include anaphylaxis (very rare), hepatic eosinophilic infiltration, and autoimmune vasculitis [133].
Churg–Strauss Syndrome (CSS), also known as allergic granulomatous angiitis, has been reported in adult patients, and no cases seem to have been reported in children [161,162]. MTK treatment has been associated with a 7.5-fold higher risk of developing CSS [163,164,165,166]. This syndrome is a rare vasculitis disorder of small- and medium-size vessels and could be characterized by blood eosinophilia and eosinophilic infiltration into affected tissues [167]. Patients who experienced CSS developed eosinophilia, leucocytosis, pulmonary infiltrates, malaise, fever, rash, neuropathy, and biomarker alterations (e.g., antineutrophil cytoplasmatic antibody and serum bilirubin) [168,169,170,171,172,173,174,175,176].
Henoch–Schlönlein syndrome affects mainly male children between 3 and 15 years old and is characterized by a tetrad of clinical manifestations including palpable purpura, arthritis–arthralgia, abdominal pain, and renal disease [177]. All secondary effects disappeared on MTK removal.

3.2.5. Montelukast Administration during Pregnancy

Maternal asthma has been associated with an increased risk of pregnancy complications, including pre-eclampsia, vaginal haemorrhage, pregnancy-induced hypertension, and low birth weight [178]. Currently, montelukast is classified as a category B drug in pregnancy risk information (no evidence of risk is associated), with limited information available.
Despite the identification of limb reduction defects in live-born offspring from mothers treated with MTK [179] during the post-marketing surveillance phase by Merck (1997–2006), there are not enough studies to support or refute the possible causes of these observations. In fact, several studies claimed no association between MTK and teratogenicity or risks of adverse prenatal outcomes [179,180,181]. Since preterm birth and maternal complications (preeclampsia and gestational diabetes) are also associated with asthma [182], conclusions regarding MTK safety during pregnancy should be interpreted carefully.
To summarize, neuropsychiatric adverse side effects are the most reported ones and occur mainly in children. Hepatotoxicity should also be monitored with care, as well as the possible occurrence of immune responses after montelukast exposure.

3.3. Montelukast Repurposing Applications

Recently, montelukast has been proposed for repurposing in other therapeutic applications (Table 2), with several of these potential uses already undergoing clinical trials. A mid-2021 search of the NIH Clinical Trials Database (clinicaltrials.gov) identified 29 clinical trials using MTK in the treatment of various pathologies such as bronchiolitis, osteoarthritis, rheumatoid arthritis, pain, Alzheimer’s disease, obesity and diabetes, steatohepatitis, and dengue.
Concerning the central nervous system, MTK has been suggested as a potential drug against some neurogenerative disorders, including Alzheimer’s disease [47,105,183], Parkinson’s disease [82,107,111,184,185], and Huntington’s disease [186]. Montelukast seems to be able to improve cognitive and neurological functions due to its modulation role in the inflammatory and apoptotic cascades involved in neurodegenerative features, particularly those where TNF-α, NF-κB, caspase-3, Bcl-2, MAPK, and IL-1β participate. These are among the most relevant signalling proteins involved in neurodegeneration.
Additionally, MTK also appears to lead to a decrease in α-synuclein load and in Aβ142-induced neurotoxicity. MTK has also been found to modulate the oxidative stress associated with a dysregulation of the GSH/GSSG balance or of superoxide dismutase activity, two key factors in the maintenance of redox homeostasis [47,82,105,107,111,183,186,187,188,189,190].
The application of MTK as a chemopreventive and adjuvant agent in cancer therapy has been suggested by different research teams [191,192,193,194,195]. Previous results show that MTK is able to induce cancer cell death by inhibiting cell proliferation, downregulating Bcl-2, and promoting nuclear translocation of the apoptosis-inducing factor (AIF) [191]. The downregulation of the hypoxia-inducible factor-1α (HIF-1α) [193] has also been mentioned as a mechanism targeting cancer cells, as well as the inhibition of the TNF-α-dependent IL-8 expression and the suppression of the NF-kB p65-associated histone acetyltransferase activity (HAT) activity [195].
During the COVID-19 pandemic, MTK was used as an off-label drug in the prevention and treatment of pulmonary distress in patients infected with SARS-CoV-2. Its properties as an anti-inflammatory drug allied to cardiovascular benefits on thrombosis and vascular damage, as well as the potential beneficial effects on brain functions, make this drug a good candidate against COVID-19 symptoms [196,197,198,199,200].
Table 2 summarizes the repurposing applications that have been published regarding the use of montelukast in pathologies other than asthma and allergic rhinitis. Although new applications include conditions such as cancer, cardiovascular diseases, and neurodegenerative disorders, proposed applications involving CNS pathologies represent more than 50% of the available data.
Table 2. Repurposing applications proposed for montelukast. Montelukast has been proposed for repurposing in different therapeutic applications, including cancer, degenerative disorders, and renal failure.
Table 2. Repurposing applications proposed for montelukast. Montelukast has been proposed for repurposing in different therapeutic applications, including cancer, degenerative disorders, and renal failure.
ModelsModulationOutcome
Bones and joints
C57B/6 mice with a femoral fracturePharmacological treatment with MTK↑ chondrocyte proliferation and early bone formation[201]
In vitro osteoarthritis model with chondrocytes (ATDC5)Pharmacological treatment with MTK↓ cartilage degradation; ↓ cell injury, oxidative stress, apoptosis; ↓ CysLTR1 expression; ↑ KLF2 expression[202]
Cancer
Nationwide population-based study with data from the Taiwan National Health Insurance Research DatabaseCancer patients with diagnosed asthma, treated with leukotriene inhibitors↓ cancer risk[192]
Human lung cancer cells and Lewis lung-carcinoma-bearing micePharmacological treatment with MTKCell proliferation inhibition; ↓ Bcl-2; ↑ Bak; ↑ nuclear translocation of AIF;
↓ phosphorylation of WNK1, Akt, Erk1/2, MEK, and PRAS40 proteins
[191]
Prostate cancer cell linesPharmacological treatment with MTK↓ HIF-1α protein;
↑ phosphorylation of eIF-2α
[193]
Phorbol-myristate--acetate-differentiated U937 cellsPharmacological effect of
MTK
↓ TNF-α-stimulated IL-8 expression;
no effect on NF-kB p65 activation; suppressed NF-kB p65-associated HAT activity
[195]
Tumour specimens from patients with prostate cancer and prostate cancer cell linesPharmacological treatment with MTKCysLTR1 overexpressed in prostate tissues; ↑ apoptosis of prostate cancer cells[194]
Cardiovascular
Nationwide population-based study (Swedish population)Association between MTK use and cardiovascular outcomes↓ recurrent cardiovascular events[203]
Nationwide population-based study (Swedish population)Association between MTK use and cardiovascular outcomes↓ risk of aortic stenosis[204]
Asthmatic patientsPharmacological effect of MTK on cardiovascular risk↓ levels of cardiovascular disease-associated inflammatory biomarkers and lipid levels[205]
CNS: Alzheimer’s disease
Transgenic
5xFAD Mice (AD mouse model)
Pharmacological effect of MTK on neuroinflammation (microglia and CD8+ T cells)↑ Tmem119+; ↓ genes related to AD-associated microglia; ↓ infiltration of CD8+ T-cells into the brain parenchyma; ↑ cognitive functions;
↓ 1061 genes (e.g., Gpr17, Entpd1, Mlec); ↑ 744 genes (e.g., Zfp46, Ciart, Dbp);
more pronounced effect in females
[189]
Transgenic DCX-DsRed2 and wildtype Fisher 344 rats, FoxO1/3/4fl mice, andGPR17_/_GFPmicePharmacological treatment with MTK↑ learning and memory in old rats; no effect on learning in young rats; ↓ microglia inflammation; ↑ BBB integrity; ↑ hippocampal neurogenesis; ↓ GPR17; ↓ CD68; ↑ claudin-5; ↑ PCNA, DCX, NeuN[47]
Intracerebroventricular infusions of aggregated Aβ1–42 in ICR miceRescue effect of MTK on Aβ1–42-induced neurotoxicity↓ memory impairment; ↓ inflammation and apoptosis markers;
↓ CysLTR1 mRNA/protein; ↓ IL-1β, TNF-α, NF-κB p65; ↓ caspase-3; ↑ Bcl-2
[105]
Primary mouse neurons (foetal ICR mice) treated with Aβ1–42Rescue effect of MTK on Aβ1–42-induced neurotoxicity↑ cell viability;
↓ CysLTR1 mRNA/protein; ↓ IL-1β, TNF-α; NF-κB p65; ↓ caspase-3; ↑ Bcl-2
[183]
Intracerebroventricular streptozotocin-induced model of sporadic AD in ICR micePharmacological treatment with MTK↓ memory impairment; ↓ neuroinflammation and apoptosis;
↓ CysLTR1 expression; ↓ TNF-α, IL-1β, NF-κB p65; ↓ cleaved caspase-3;
↑ Bcl-2/Bax ratio
[190]
CNS: Anti-nociception
Local antinociception model of painPharmacological treatment with MTK↓ local pain behaviour in both phases (neurogenic and inflammatory);
Involvement of L-Arg/NO/cGMP/KATP channel pathway and PPARγ receptors
[206]
CNS: Brain ischemia
Middle cerebral artery occlusion model in mice and ratsPharmacological treatment with MTK↓ behavioural dysfunction, brain infarct volume, brain atrophy, and neuron loss[207]
Bilateral carotid artery occlusion model in ratsPharmacological prophylaxis and treatment with MTK↓ oxidative stress, inflammatory and apoptotic markers (myeloperoxidase, NF-κB, TNF-α, and IL-6); ↓ glutamate and lactate dehydrogenase[83]
CNS: Dementia with Lewy bodies
Human brain specimen and female transgenic mice expressing human wild-type α-synuclein vs. their wild-type litter matesPharmacological treatment with MTK↑ memory function;
↓ α-synuclein load in the dentate gyrus; ↑ Beclin-1 expression;
autophagy as a possible mechanistic pathway
[187]
CNS: Epilepsy
Epilepsy-induced spontaneous recurrent seizures with pentylenetetrazole (PTZ) in micePharmacological treatment with MTKPrevention of PTZ-induced BBB disruption; ↓ recurrent seizures; ↓ mean amplitude of electroencephalography recording during seizures[208]
↓ recurrent seizures; ↓ frequency of daily seizures[209]
Pilocarpine-induced seizures in mice
Electrically-induced seizures in mice
CNS: Huntington’s disease
Intrastriatal-quinolinic-acid-and malonic-acid-induced Huntington’s-like symptoms in ratsPharmacological treatment with MTK↓ behavioural alterations; ↓ oxidative stress; ↓ mitochondrial dysfunction; ↓ TNF-α level[186]
CNS: Multiple Sclerosis
MOG35-55-induced experimental autoimmune encephalomyelitis in female micePharmacological treatment with MTK↓ CNS infiltration of inflammatory cells; ↓ clinical symptoms;
↓ IL-17; ↓ BBB disruption
[210]
CNS: neurological ageing
Observational study using data from two databases: NorPD and the Tromsø StudyAssociation between MTK use and neurological healthImproved cognitive and neurologic function[188]
CNS: Parkinson’s disease
Rotenone-induced model of PD in ratsPharmacological treatment with MTK↑ locomotor activity; ↓ immobility time; ↓brain MDA levels; ↑ GSH levels; ↓ TNF-α levels[111]
↑ locomotor activity;
↓ p38 MAPK, TNF-α, IL-1β, NF-κB; ↓ CysLTR1 expression; ↓ p53 mRNA, caspase-3;
↑ GSH, SOD; ↓ MDA levels
[82]
6-Hydroxydopamine mouse model (C57BL/6 mice) of PDTherapeutic effects of MTK↓ TNF-α levels; ↓ IL-1β[107]
COVID-19
Computational methodsTarget-based virtual ligand screening and molecular dockingWell-fitted in the active pocket of SARS-CoV-2 3CLpro, Mpro and RdRp[211,212]
Retrospective study of COVID patientsCOVID patients treated with or without MTK↓ events of clinical deterioration[213]
Glaucoma
Magnetic microbead injection into the anterior chamber of female Brown Norway ratsPharmacological treatment with MTK↓ intra ocular pressure; ↑ retinal ganglion cell survival in ocular hypertension eyes; ↓ activation of Iba1+ microglial cells in retina; ↓ GPR17+ cells[214]
Lung transplant
Bronchiolitis obliterans syndrome after lung transplantation in patientsPharmacological treatment with MTK↓ forced expiratory volume in 1 s (FEV1)[215,216,217]
Pulmonary fibrosis
Bleomycin-induced pulmonary fibrosis in female C57BL/6J micePharmacological prophylaxis and treatment with MTK↓ fibrotic area; ↓ IL-6, IL-10, IL-13, and TGF-β1 mRNA levels; ↑ CysLTR2 mRNA expression[218]
Renal failure
Rhabdomyolysis-induced acute renal failure in Wistar ratsPharmacological prophylaxis and treatment with MTKImproved functional and structural renal damage; ↓ tubular damage;
↓ serum creatinine and urea levels; ↓ serum phosphate levels; ↓ GSH and MDA levels; ↑ SOD levels; ↓ serum TNF-α, TGF-β1, Fas, IL-10; ↑ IL-6/ TNF-α ratio
[219]
Cisplatin-induced renal dysfunction in male Sprague Dawley ratsPharmacological prophylaxis and treatment with MTKAmeliorated renal toxicity;
↓ responsiveness to acetylcholine;
↓ serum creatinine, blood urea nitrogen, LDH; ↑ serum albumin to normal levels; ↑ GSH levels; ↓ SOD levels
[220]
Pyelonephritis induced by Escherichia coli in Wistar ratsPharmacological treatment with MTK↓ severity of kidney damage and renal scarring; ↓ serum TNF-α, creatinine, blood urea nitrogen, MDA levels; ↑ GSH levels[221]
According to Table 2, MTK is a very promising drug that shows a lot of potential in various disorders. Nowadays, the strategy of drug repurposing is being followed by many pharmaceutical companies [222] since it allows a faster and less cost-effective drug development, with the skipping of some development steps and clinical trial phases. In fact, some of the more known instances of drug repurposing and interest in off-target drug effects were the use of drugs in the treatment of COVID-19 symptoms during the pandemic, which included MTK [213]. Additional examples are the repurposing of minoxidil (an antihypertensive agent) as hair growth stimulant, and the use of sildenafil (developed for angina) in erectile dysfunction or pulmonary hypertension [222].
As MTK has been on the market since 1998, some of its proposed repurposing applications were based on retrospective studies, whereas others resulted from ADRs or serendipity [188,192,203,204]. Considering all the findings until now (clinical and in vitro/in vivo), it is critical to unveil the mechanisms involved in these new potential applications, in order to validate the repurposing uses. Once validated, patients could benefit from a potential new drug that will contribute to a better management of their symptoms, improving their quality of life. At present, most applications are based on the wide anti-inflammatory properties of MTK.

4. Human Neurodegenerative Diseases

Neurodegenerative disorders affect millions of people around the world and show increasing prevalence. These disorders are caused by the progressive degeneration and/or loss of a specific neuron population due to chronic neuroinflammation. The most common disorders are amyloidoses, tauopathies, α-synucleinopathies, and transactivation response DNA binding protein 43 (TDP-43) proteinopathies, which may cause movement or functional problems, such as Alzheimer’s disease (AD), Parkinson’s disease (PD), Lewy body disorders, and amyotrophic lateral sclerosis, among others [223].
Autopsies of older people have shown that aged brains develop abnormal accumulation of hyperphosphorylated Tau protein, amyloid-β deposits, accumulation of TDP-43, and α-synuclein deposits. Brains from people aged 90 and older have lost around 11% of their weight (approximately 150 g of brain tissue) when compared with people in their fifties. This weight decrease could be related to the loss of neurons and glia cells, myelin, fluid, or other factors. Despite the observation of these physiological changes in the aged brain, not all autopsied people suffered from neurodegenerative diseases, which suggests that some people might have compensatory mechanisms that enable them to maintain normal cognition [224,225]. During normal (healthy) ageing, intact synapses are maintained by APP processing through a non-amyloidogenic pathway, with amyloid production being balanced by clearance processes. The APP protein, as well as products resulting from its processing, play an important role in functions such as synaptogenesis, axonal growth, synaptic plasticity, learning, and memory. Furthermore, the Tau protein is involved in neuronal microtubule stabilization [226].
This section will further focus on two neurodegenerative diseases: Alzheimer’s and Parkinson’s diseases.

4.1. Alzheimer’s Disease

Nowadays, 47 million people suffer from dementia in the world, and it is expected that this number will increase to 131 million patients by 2050 [227]. Among these, around 80% of dementia is caused by Alzheimer’s disease [227,228]. AD is a progressive disease whose pathological changes start decades before the clinical symptoms, leading to the development of cognitive impairment, functional symptoms, and, later, dementia [227,228].
AD is classified in two forms: the familial early-onset (FAD) form and the sporadic late-onset (SAD) form. The FAD form affects approximately 5% of AD patients and is diagnosed in individuals between 30 and 60 years of age [67]. These patients present hereditary mutations in several genes involved in Aβ formations, such as the genes encoding β amyloid precursor protein (APP) and presenilin 1 and/or 2, which contribute to the early onset of symptoms [67]. The SAD form is mostly associated with age (patients older than 65 years of age), and the risk factors include pathways involved in cholesterol metabolism (APOE, CLU, and ABCA7), immune response (CR1, CD33, and Trem2), and endocytosis (PICALM and EPHA1, and ED2AP) [67,229,230,231,232,233].
AD is characterized by two major pathological hallmarks, namely the accumulation of β-amyloid (Aβ) plaques outside neurons and the accumulation of phosphorylated Tau protein, also known as neurofibrillary tangles (NFT), inside and outside neurons due to hyperphosphorylated Tau protein aggregation. The instability and reduced axonal transport caused by the loss of Tau function, as well as the formation of amyloid plaques, leads to damage and disruption of neuronal synapses and, later, cell death [234] (Figure 6B). Additionally, AD patients also display loss of neurons and white matter (brain atrophy and neurodegeneration), cerebral amyloid angiopathy (accumulation of amyloid plaques in the leptomeninges and small/medium-sized cerebral blood vessels, leading to fragile vessels), neuroinflammation, oxidative damage, and neurotransmitter imbalance [67,235,236].
The Aβ peptide is expressed as part of a 695-amino-acid polypeptide, the amyloid-β precursor protein, which is a glycosylated transmembrane protein encoded by a gene located on human chromosome 21 [237,238]. APP can be processed through two alternative pathways (Figure 6C): a non-amyloidogenic pathway, where APP is firstly cleaved by α-secretase, and an amyloidogenic pathway where APP cleavage is performed by β-secretase.
In the non-amyloidogenic pathway, α-secretase catalyses the release of a soluble amyloid precursor protein α (sAPPα) and an α-C-terminal fragment (CTFα or C83); the latter is converted by γ-secretase into an extracellular P3 peptide and an APP intracellular domain (AICD) peptide [239,240].
In the amyloidogenic pathway, β-secretase catalyses the formation of a β-C-terminal fragment (CTFβ or C99) and an N-terminal sAPPβ fragment. CTFβ is then cleaved by γ-secretase, releasing extracellular Aβ peptides of different length and the APP intracellular domain [239,240]. The most frequent final Aβ forms are the 40- (Aβ40) and 42-amino acid (Aβ42) peptides [240]. Aβ42 is more neurotoxic than Aβ40, possibly due to its higher tendency to produce oligomers [241].
Figure 6. The molecular basis of Alzheimer’s disease. (A) Healthy cognitive ageing. (B) Alzheimer’s disease pathology. (C) Human APP proteolytic pathways. Adapted from Müller et al. [226], De Strooper et al. [239], Chen et al. [240]. Created with BioRender.com.
Figure 6. The molecular basis of Alzheimer’s disease. (A) Healthy cognitive ageing. (B) Alzheimer’s disease pathology. (C) Human APP proteolytic pathways. Adapted from Müller et al. [226], De Strooper et al. [239], Chen et al. [240]. Created with BioRender.com.
Pharmaceuticals 15 01039 g006
The accumulation of Aβ is caused by an imbalance between production, clearance (or degradation), and aggregation of peptides [236]. Whereas mutations in APP, Psen1 and/or Psen2 genes are associated with Aβ processing in FAD [242], SAD is linked to an accumulation of Aβ caused by a decrease in clearance mechanisms [243]. Clearance can occur within the brain or after transport from the brain to the periphery (liver and kidney) and includes proteolytic pathways that depend on neprilysin (NE), insulin-degrading enzyme (IDE), matrix metalloproteinases (MMPs), angiotensin-converting enzyme (ACE), endothelin-converting enzyme (ECE), plasmin, the activity of the ubiquitin–proteosome system, the autophagy–lysosome system, or microglial phagocytosis [243,244].
In addition to the characteristic Aβ peptide plaques and to phosphorylated Tau accumulation, the brain of some AD patients contains high levels of α-synuclein. This protein is expressed in kidney, blood cells, and, predominantly, in neurons; it is mainly known for its association with Lewy bodies and Parkinson’s disease pathologies. When present in high levels, α-synuclein forms oligomers and fibrils. Moreover, recent studies have suggested that amyloid plaques are able to promote the formation of α-synuclein aggregates, increasing neurotoxicity [245,246].
Currently, no treatments are available for Alzheimer’s disease. However, some drugs that temporarily improve disease symptoms are currently used: rivastigmine, galantamine, and donepezil, which are cholinesterase inhibitors, increasing neurotransmitter levels in the brain; and memantine, an antagonist of the N-methyl-D-aspartate receptor. Recently, aducanumab, a monoclonal antibody targeting the N-terminal pyroglutamate Aβ epitope, which could help in the reduction of the amyloid plaque level, was the first drug of this class to be approved by the US FDA [240,247,248].

4.2. Parkinson’s Disease

According to the British National Health Service (NHS), 1 in 500 people suffer from Parkinson’s disease. This neurodegenerative disorder is mainly characterized by the loss of dopaminergic neurons in the midbrain substantia nigra pars compacta region, which causes motor and nonmotor symptoms. The motor symptoms, collectively known as parkinsonism, include bradycardia, rigidity, resting tremor, and impairment of postural balance. Nonmotor symptoms include depression, anxiety, sleep disturbance, constipation, dementia, and cognitive decline [89,249,250].
As mentioned, the hallmark of PD is the progressive loss of dopaminergic neurons present in the substantia nigra, with the appearance of Lewy neurites and Lewy bodies—intracellular inclusions of α-synuclein aggregates—that ultimately lead to dopaminergic neuron death [249,250]. In addition to α-synuclein, Lewy bodies can also contain misfolded phosphorylated Tau and Aβ proteins, increasing neuronal toxicity [250]. Thus, the mechanisms involved in Parkinson’s disease pathology include the accumulation of misfolded protein aggregates, loss of protein clearance mechanisms, mitochondrial damage, oxidative stress, excitotoxicity, and neuroinflammation [250].
Besides dopaminergic neurons, serotoninergic neurons are also involved in PD. Dysfunctions in serotoninergic neurotransmission contribute to motor and nonmotor symptoms, such as resting tremor, dyskinesia, depression, and anxiety [251,252].
Currently, no treatment is available for Parkinson’s disease and the prescribed drugs only allow the control of some symptoms: levodopa (a precursor of dopamine), dopamine receptor agonists, inhibitors of monoamine oxidase B (MAO-B), catechol-O-methyltransferase (COMT) inhibitors, amantadine (an anti-influenza drug widely used in parkinsonism and dyskinesia), and anticholinergic drugs, including antidepressant drugs [89]. The use of MAO-B inhibitors intends not only to decrease the metabolism of neurotransmitters such as dopamine, increasing their extracellular concentration, but also to reduce the oxidative stress produced by MAO-B activity [253].
MTK repurposing for AD and PD management has been investigated in several works, using various biological systems, from cell-based systems to transgenic animals, as reviewed in Table 2. Most of the published studies indicate that MTK administration leads to a diminished inflammatory status at the CNS level, in agreement with MTK actions as CysLTR1 antagonist and with the contribution of neuroinflammatory glial processes to neurodegeneration progress. However, and until the mechanisms underlying MTK’s toxicity are fully elucidated, any potential applications of MTK beyond the officially approved ones must be considered with extreme care.

5. Conclusions

MTK is a widely used leukotriene antagonist drug, targeted at asthma management for adults and children alike. Regarding lactation, MTK levels found in human milk were below therapeutic ranges established for children, supporting the safety of this treatment for asthmatic breastfeeding mothers [254].
MTK’s anti-inflammatory action is not confined to the respiratory system but is of a more systemic nature, which has led to the development of clinical studies aiming at MTK repurposing for various inflammatory-based conditions, particularly aiming at the management of a number of neurodegenerative diseases.
However, MTK is associated with a number of adverse drug reactions, particularly at the CNS level, where neuropsychiatric events are linked to MTK administration but promptly resolve upon stopping the treatment. Although the molecular basis for these toxic side effects is still unknown, they must be taken into consideration when addressing the potential repurposing of MTK.
The metabolism of MTK has been studied extensively by various authors, as reviewed above. A link between metabolism and toxicity was not identified from the various observed phase I and phase II metabolites, indicating that MTK’s toxicity is likely mediated via interaction with biological pathways and not through chemical reaction with biomolecules.

Author Contributions

Conceptualization, C.F.M.; writing—original draft preparation, C.F.M.; writing—review and editing, M.M.M. and G.C.J. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Fundação para a Ciência e a Tecnologia (FCT), grant numbers SAICTPAC/0019/2015 (M.M.M.), PTDC/QUI-QAN/32242/2017 (M.M.M. and G.C.J.), and RNEM-LISBOA-01-0145-FEDER-022125 (Portuguese Mass Spectrometry Network). C.F.M. acknowledges a PhD fellowship from Fundação para a Ciência e a Tecnologia (PD/BD/143128/2019 and COVID/BD/152559/2022). Centro de Química Estrutural is a Research Unit funded by FCT through projects UIDB/00100/2020 and UIDP/00100/2020. Institute of Molecular Sciences is an Associate Laboratory funded by FCT through project LA/P/0056/2020.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

3CLpro3-Chymotrypsin-like protease
5-HpETE5(S)-Hydroperoxyeicosatetraenoic acid
5-LOX5-Lipoxygenase
AAArachidonic acid
ADAlzheimer’s disease
ADRsAdverse drug reactions
AIFApoptosis-inducing factor
AktProtein kinase B
APPAmyloid-beta precursor protein
β-Amyloid protein
1–42β-Amyloid peptide, amino acids 1 to 42
B-LTLeukotriene B receptors
BakBcl-2 homologous antagonist/killer
BAXBcl-2-associated X protein
BBBBlood–brain barrier
Bcl-2B-cell lymphoma 2
CCL2C-C motif chemokine 2
CDCluster of differentiation
cGMP/KATPCyclic monophosphate/ATP-sensitive potassium
CiartCircadian associated repressor of transcription
CNSCentral nervous system
COVID-19SARS-CoV-2 disease
cPLA2Cytosolic phospholipase A2
CSSChurg–Strauss Syndrome
CTFC terminal fragment
CYPCytochrome P450
CysLTRCysteinyl leukotrienes receptor (isoforms 1, 2, and 3)
CysLTsCysteinyl leukotrienes
DbpD site albumin promoter binding protein
DCXDoublecortin
DPDipeptidase
eIF-2αEukaryotic initiation factor-2α
Entpd1Ectonucleoside triphosphate diphosphohydrolase 1
Erk1/2Extracellular signal-regulated kinase 1/2
FADFamilial early-onset Alzheimer’s disease
FasTumour necrosis factor receptor superfamily member 6
FLAP5-LOX activating protein
GGLTγ-Glutamyl leukotrienase
GGTγ-Glutamyl transpeptidase
GPR17G Protein-Coupled Receptor 17
GSHGlutathione
GSSGGlutathione disulphide
HATHistone acetyltransferase
HDHungtinton’s disease
HIF-1αHypoxia-inducible factor-1
Iba1Ionized calcium-binding adaptor molecule 1
IFN-γInterferon-γ
ILInterleukin
iNOSInducible nitric oxide synthase
KLF2Krüppel-like Factor 2
LDHLactate dehydrogenase
LPSLipopolysaccharide
LTLeukotriene
LTA4Leukotriene A4
LTA4HLeukotriene A4 hydrolase
LTB4Leukotriene B4
LTC4Leukotriene C4
LTC4SLeukotriene C4 synthase
LTD4Leukotriene D4
LTE4Leukotriene E4
MAPKMitogen-activated protein kinase
MDAMalondialdehyde
MEKExtracellular signal-regulated kinase kinase
MlecMalectin protein
MOGMyelin oligodendrocyte glycoprotein
MproSARS-CoV-2 Main protease
mRNAMessenger RNA
MRPMultidrug resistance proteins (isoforms 1 and 4)
MTKMontelukast
NeuNNeuronal nuclear protein
NF-κBNuclear factor kappa B
P2Y12P2Y purinoceptor 12
p38 MAPKp38 mitogen-activated protein kinase
PCNAProliferating cell nuclear antigen
PDParkinson’s disease
PGProstaglandin
PPAR-αPeroxisomal proliferator-activated receptor α
PPARγProliferator-activated receptor γ
PRAS40Proline-rich Akt substrate of 40 kDa
PsenPresenilin
PTZPentylenetetrazole
RdRpRNA dependent RNA polymerase
ROSReactive oxygen species
SADSporadic late-onset Alzheimer’s disease
SARS-CoV-2Severe acute respiratory syndrome coronavirus 2
SMCSmooth muscle cells
SODSuperoxide dismutase
TGF-β1Transforming growth factor-beta 1
Tmem119Transmembrane protein 119
TNF-αTumour necrosis factor α
UGTGlucuronosyltransferase
WNK1WNK lysine deficient protein kinase 1
Zfp46Zinc finger protein 46

References

  1. Bäck, M. Leukotriene signaling in atherosclerosis and ischemia. Cardiovasc. Drugs Ther. 2009, 23, 41–48. [Google Scholar] [CrossRef] [PubMed]
  2. Brocklehurst, W.E. The release of histamine and formation of a slow-reacting substance (SRS-A) during anaphylactic shock. J. Physiol. 1960, 151, 416–435. [Google Scholar] [CrossRef]
  3. Rius, M.; Hummel-Eisenbeiss, J.; Keppler, D. ATP-dependent transport of leukotrienes B4 and C4 by the multidrug resistance protein ABCC4 (MRP4). J. Pharmacol. Exp. Ther. 2008, 324, 86–94. [Google Scholar] [CrossRef] [PubMed]
  4. Johnson, Z.L.; Chen, J. Structural Basis of Substrate Recognition by the Multidrug Resistance Protein MRP1. Cell 2017, 168, 1075–1085.e9. [Google Scholar] [CrossRef]
  5. Samuelsson, B.; Dáhlen, S.E.; Lindgren, J.A.; Rouzer, C.A.; Serhan, C.N. Leukotrienes and Lipoxins—Structures, Biosynthesis, and Biological Effects. Science 1987, 237, 1171–1176. [Google Scholar] [CrossRef] [PubMed]
  6. Heidenreich, K.A.; Corser-Jensen, C.E. Chapter 12-5-Lipoxygenase-Activating Protein Inhibitors: Promising Drugs for Treating Acute and Chronic Neuroinflammation Following Brain Injury. In New Therapeutics for Traumatic Brain Injury; Heidenreich, K.A., Ed.; Academic Press: San Diego, CA, USA, 2017; pp. 199–210. [Google Scholar]
  7. Dennis, E.A.; Norris, P.C. Eicosanoid storm in infection and inflammation. Nat. Rev. Immunol. 2015, 15, 511–523. [Google Scholar] [CrossRef]
  8. Massoumi, R.; Sjölander, A. The role of leukotriene receptor signaling in inflammation and cancer. Sci. World J. 2007, 7, 1413–1421. [Google Scholar] [CrossRef]
  9. Kanaoka, Y.; Boyce, J.A. Cysteinyl leukotrienes and their receptors: Cellular distribution and function in immune and inflammatory responses. J. Immunol. 2004, 173, 1503–1510. [Google Scholar] [CrossRef]
  10. Capra, V. Molecular and functional aspects of human cysteinyl leukotriene receptors. Pharm. Res. 2004, 50, 1–11. [Google Scholar] [CrossRef]
  11. Singh, R.K.; Gupta, S.; Dastidar, S.; Ray, A. Cysteinyl leukotrienes and their receptors: Molecular and functional characteristics. Pharmacology 2010, 85, 336–349. [Google Scholar] [CrossRef]
  12. Singh, R.K.; Tandon, R.; Dastidar, S.G.; Ray, A. A review on leukotrienes and their receptors with reference to asthma. J. Asthma. 2013, 50, 922–931. [Google Scholar] [CrossRef] [PubMed]
  13. Liu, F.; Ouyang, J.; Sharma, A.N.; Liu, S.; Yang, B.; Xiong, W.; Xu, R. Leukotriene inhibitors for bronchiolitis in infants and young children. Cochrane Database Syst. Rev. 2015. [Google Scholar] [CrossRef] [PubMed]
  14. Yokomizo, T.; Nakamura, M.; Shimizu, T. Leukotriene receptors as potential therapeutic targets. J. Clin. Investig. 2018, 128, 2691–2701. [Google Scholar] [CrossRef]
  15. Smith, W.L.; Murphy, R.C. The Eicosanoids. In Biochemistry of Lipids, Lipoproteins and Membranes, 6th ed.; Ridgway, N.D., McLeod, R.S., Eds.; Academic Press: Cambridge, MA, USA, 2016; pp. 259–296. [Google Scholar]
  16. Bäck, M. Leukotrienes: Potential therapeutic targets in cardiovascular diseases. Bull. Acad. Natl. Med. 2006, 190, 1511–1521. [Google Scholar]
  17. Bäck, M. Inhibitors of the 5-lipoxygenase pathway in atherosclerosis. Curr. Pharm. Des. 2009, 15, 3116–3132. [Google Scholar] [CrossRef] [PubMed]
  18. Nakamura, M.; Shimizu, T. Leukotriene receptors. Chem. Rev. 2011, 111, 6231–6298. [Google Scholar] [CrossRef] [PubMed]
  19. Funk, C.D. Prostaglandins and leukotrienes: Advances in eicosanoid biology. Science 2001, 294, 1871–1875. [Google Scholar] [CrossRef]
  20. Capra, V.; Accomazzo, M.R.; Gardoni, F.; Barbieri, S.; Rovati, G.E. A role for inflammatory mediators in heterologous desensitization of CysLT1 receptor in human monocytes. J. Lipid. Res. 2010, 51, 1075–1084. [Google Scholar] [CrossRef] [PubMed]
  21. Liu, M.; Yokomizo, T. The role of leukotrienes in allergic diseases. Allergol. Int. 2015, 64, 17–26. [Google Scholar] [CrossRef]
  22. Wunder, F.; Tinel, H.; Kast, R.; Geerts, A.; Becker, E.M.; Kolkhof, P.; Hutter, J.; Erguden, J.; Härter, M. Pharmacological characterization of the first potent and selective antagonist at the cysteinyl leukotriene 2 (CysLT2) receptor. Br. J. Pharmacol. 2010, 160, 399–409. [Google Scholar] [CrossRef]
  23. Poff, C.D.; Balazy, M. Drugs that target lipoxygenases and leukotrienes as emerging therapies for asthma and cancer. Curr. Drug Targets Inflamm. Allergy 2004, 3, 19–33. [Google Scholar] [CrossRef] [PubMed]
  24. Gauvreau, G.M.; Boulet, L.P.; FitzGerald, J.M.; Cockcroft, D.W.; Davis, B.E.; Leigh, R.; Tanaka, M.; Fourre, J.A.; Tanaka, M.; Nabata, T.; et al. A dual CysLT1/2 antagonist attenuates allergen-induced airway responses in subjects with mild allergic asthma. Allergy 2016, 71, 1721–1727. [Google Scholar] [CrossRef] [PubMed]
  25. Itadani, S.; Yashiro, K.; Aratani, Y.; Sekiguchi, T.; Kinoshita, A.; Moriguchi, H.; Ohta, N.; Takahashi, S.; Ishida, A.; Tajima, Y.; et al. Discovery of Gemilukast (ONO-6950), a Dual CysLT1 and CysLT2 Antagonist As a Therapeutic Agent for Asthma. J. Med. Chem. 2015, 58, 6093–6113. [Google Scholar] [CrossRef] [PubMed]
  26. Lynch, K.R.; O'Neill, G.P.; Liu, Q.; Im, D.S.; Sawyer, N.; Metters, K.M.; Coulombe, N.; Abramovitz, M.; Figueroa, D.J.; Zeng, Z.; et al. Characterization of the human cysteinyl leukotriene CysLT1 receptor. Nature 1999, 399, 789–793. [Google Scholar] [CrossRef]
  27. Heise, C.E.; O'Dowd, B.F.; Figueroa, D.J.; Sawyer, N.; Nguyen, T.; Im, D.S.; Stocco, R.; Bellefeuille, J.N.; Abramovitz, M.; Cheng, R.; et al. Characterization of the human cysteinyl leukotriene 2 receptor. J. Biol. Chem. 2000, 275, 30531–30536. [Google Scholar] [CrossRef]
  28. Sarau, H.M.; Ames, R.S.; Chambers, J.; Ellis, C.; Elshourbagy, N.; Foley, J.J.; Schmidt, D.B.; Muccitelli, R.M.; Jenkins, O.; Murdock, P.R.; et al. Identification, molecular cloning, expression, and characterization of a cysteinyl leukotriene receptor. Mol. Pharmacol. 1999, 56, 657–663. [Google Scholar] [CrossRef]
  29. J Jones, T.R.; Zamboni, R.; Belley, M.; Champion, E.; Charette, L.; Ford-Hutchinson, A.W.; Frenette, R.; Gauthier, J.Y.; Leger, S.; Masson, P.; et al. Pharmacology of L-660,711 (MK-571): A novel potent and selective leukotriene D4 receptor antagonist. Can. J. Physiol. Pharmacol. 1989, 67, 17–28. [Google Scholar] [CrossRef]
  30. Ni, N.C.; Yan, D.; Ballantyne, L.L.; Barajas-Espinosa, A.; St Amand, T.; Pratt, D.A.; Funk, C.D. A selective cysteinyl leukotriene receptor 2 antagonist blocks myocardial ischemia/reperfusion injury and vascular permeability in mice. J. Pharmacol. Exp. Ther. 2011, 339, 768–778. [Google Scholar] [CrossRef]
  31. Abbracchio, M.P.; Burnstock, G.; Boeynaems, J.M.; Barnard, E.A.; Boyer, J.L.; Kennedy, C.; Miras-Portugal, M.T.; King, B.F.; Gachet, C.; Jacobson, K.A.; et al. The recently deorphanized GPR80 (GPR99) proposed to be the P2Y15 receptor is not a genuine P2Y receptor. Trends Pharmacol. Sci. 2005, 26, 8–9. [Google Scholar] [CrossRef]
  32. Kanaoka, Y.; Maekawa, A.; Austen, K.F. 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. [Google Scholar] [CrossRef]
  33. Cherif, H.; Duhamel, F.; Cécyre, B.; Bouchard, A.; Quintal, A.; Chemtob, S.; Bouchard, J.F. Receptors of intermediates of carbohydrate metabolism, GPR91 and GPR99, mediate axon growth. PLoS Biol. 2018, 16, e2003619. [Google Scholar] [CrossRef] [PubMed]
  34. Rajkumar, P.; Pluznick, J.L. Unsung renal receptors: Orphan G-protein-coupled receptors play essential roles in renal development and homeostasis. Acta Physiol. (Oxf) 2017, 220, 189–200. [Google Scholar] [CrossRef] [PubMed]
  35. Wittenberger, T.; Hellebrand, S.; Munck, A.; Kreienkamp, H.J.; Schaller, H.C.; Hampe, W. GPR99, a new G protein-coupled receptor with homology to a new subgroup of nucleotide receptors. BMC Genomics. 2002, 3, 17. [Google Scholar] [CrossRef] [PubMed]
  36. Peebles, R.S. Antileukotriene Therapy in Asthma. In Middleton’s Allergy: Principles and Practice, 9th ed.; Burks, A.W., Holgate, S.T., O'Hehir, R.E., Bacharier, L.B., Broide, D.H., Hershey, G.K.K., Peebles, S., Eds.; Elsevier: Edinburgh, UK, 2020; pp. 1584–1598. [Google Scholar]
  37. Paruchuri, S.; Tashimo, H.; Feng, C.; Maekawa, A.; Xing, W.; Jiang, Y.; Kanaoka, Y.; Conley, P.; Boyce, J.A. Leukotriene E4-induced pulmonary inflammation is mediated by the P2Y12 receptor. J. Exp. Med. 2009, 206, 2543–2555. [Google Scholar] [CrossRef] [PubMed]
  38. Haynes, S.E.; Hollopeter, G.; Yang, G.; Kurpius, D.; Dailey, M.E.; Gan, W.B.; Julius, D. The P2Y12 receptor regulates microglial activation by extracellular nucleotides. Nat. Neurosci. 2006, 9, 1512–1519. [Google Scholar] [CrossRef]
  39. Moore, C.S.; Ase, A.R.; Kinsara, A.; Rao, V.T.; Michell-Robinson, M.; Leong, S.Y.; Butovsky, O.; Ludwin, S.K.; Séguéla, P.; Bar-Or, A.; et al. P2Y12 expression and function in alternatively activated human microglia. Neurol. Neuroimmunol. Neuroinflamm. 2015, 2, e80. [Google Scholar] [CrossRef]
  40. Hollopeter, G.; Jantzen, H.M.; Vincent, D.; Li, G.; England, L.; Ramakrishnan, V.; Yang, R.B.; Nurden, P.; Nurden, A.; Julius, D.; et al. Identification of the platelet ADP receptor targeted by antithrombotic drugs. Nature 2001, 409, 202–207. [Google Scholar] [CrossRef]
  41. Gómez Morillas, A.; Besson, V.C.; Lerouet, D. Microglia and Neuroinflammation: What Place for P2RY12? Int. J. Mol. Sci. 2021, 22, 1636. [Google Scholar] [CrossRef]
  42. Neves, J.S.; Radke, A.L.; Weller, P.F. Cysteinyl leukotrienes acting via granule membrane-expressed receptors elicit secretion from within cell-free human eosinophil granules. J. Allergy Clin. Immunol. 2010, 125, 477–482. [Google Scholar] [CrossRef]
  43. Suh, D.H.; Trinh, H.K.T.; Liu, J.N.; Pham, L.D.; Park, S.M.; Park, H.S.; Shin, Y.S. P2Y12 antagonist attenuates eosinophilic inflammation and airway hyperresponsiveness in a mouse model of asthma. J. Cell Mol. Med. 2016, 20, 333–341. [Google Scholar] [CrossRef]
  44. Collet, J.P.; O’Connor, S. Clinical effects and outcomes with new P2Y12 inhibitors in ACS. Fund. Clin. Pharmacol. 2012, 26, 16–18. [Google Scholar] [CrossRef] [PubMed]
  45. Fumagalli, M.; Daniele, S.; Lecca, D.; Lee, P.R.; Parravicini, C.; Fields, R.D.; Rosa, P.; Antonucci, F.; Verderio, C.; Trincavelli, M.L.; et al. Phenotypic changes, signaling pathway, and functional correlates of GPR17-expressing neural precursor cells during oligodendrocyte differentiation. J. Biol. Chem. 2011, 286, 10593–10604. [Google Scholar] [CrossRef] [PubMed]
  46. Ciana, P.; Fumagalli, M.; Trincavelli, M.L.; Verderio, C.; Rosa, P.; Lecca, D.; Ferrario, S.; Parravicini, C.; Capra, V.; Gelosa, P.; et al. The orphan receptor GPR17 identified as a new dual uracil nucleotides/cysteinyl-leukotrienes receptor. EMBO J. 2006, 25, 4615–4627. [Google Scholar] [CrossRef] [PubMed]
  47. Marschallinger, J.; Schäffner, I.; Klein, B.; Gelfert, R.; Rivera, F.J.; Illes, S.; Grassner, L.; Janssen, M.; Rotheneichner, P.; Schmuckermair, C.; et al. Structural and functional rejuvenation of the aged brain by an approved anti-asthmatic drug. Nat. Commun. 2015, 6, 8466. [Google Scholar] [CrossRef] [PubMed]
  48. Fumagalli, M.; Lecca, D.; Coppolino, G.T.; Parravicini, C.; Abbracchio, M.P. Pharmacological Properties and Biological Functions of the GPR17 Receptor, a Potential Target for Neuro-Regenerative Medicine. Adv. Exp. Med. Biol. 2017, 1051, 169–192. [Google Scholar] [CrossRef]
  49. Maekawa, A.; Xing, W.; Austen, K.F.; Kanaoka, Y. GPR17 regulates immune pulmonary inflammation induced by house dust mites. J. Immunol. 2010, 185, 1846–1854. [Google Scholar] [CrossRef]
  50. Zhao, B.; Zhao, C.Z.; Zhang, X.Y.; Huang, X.Q.; Shi, W.Z.; Fang, S.H.; Lu, Y.B.; Zhang, W.P.; Xia, Q.; Wei, E.Q. 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. [Google Scholar] [CrossRef] [PubMed]
  51. Ceruti, S.; Villa, G.; Genovese, T.; Mazzon, E.; Longhi, R.; Rosa, P.; Bramanti, P.; Cuzzocrea, S.; Abbracchio, M.P. The P2Y-like receptor GPR17 as a sensor of damage and a new potential target in spinal cord injury. Brain 2009, 132, 2206–2218. [Google Scholar] [CrossRef]
  52. Burnstock, G. An introduction to the roles of purinergic signalling in neurodegeneration, neuroprotection and neuroregeneration. Neuropharmacology 2016, 104, 4–17. [Google Scholar] [CrossRef]
  53. Franke, H.; Parravicini, C.; Lecca, D.; Zanier, E.R.; Heine, C.; Bremicker, K.; Fumagalli, M.; Rosa, P.; Longhi, L.; Stocchetti, N.; et al. Changes of the GPR17 receptor, a new target for neurorepair, in neurons and glial cells in patients with traumatic brain injury. Purinergic. Signal. 2013, 9, 451–462. [Google Scholar] [CrossRef]
  54. Lecca, D.; Trincavelli, M.L.; Gelosa, P.; Sironi, L.; Ciana, P.; Fumagalli, M.; Villa, G.; Verderio, C.; Grumelli, C.; Guerrini, U.; et al. The recently identified P2Y-like receptor GPR17 is a sensor of brain damage and a new target for brain repair. PLoS ONE 2008, 3, e3579. [Google Scholar] [CrossRef] [PubMed]
  55. Dziedzic, A.; Miller, E.; Saluk-Bijak, J.; Bijak, M. The GPR17 Receptor-A Promising Goal for Therapy and a Potential Marker of the Neurodegenerative Process in Multiple Sclerosis. Int. J. Mol. Sci. 2020, 21, 1852. [Google Scholar] [CrossRef] [PubMed]
  56. Ou, Z.; Ma, Y.; Sun, Y.; Zheng, G.; Wang, S.; Xing, R.; Chen, X.; Han, Y.; Wang, J.; Lu, Q.R.; et al. A GPR17-cAMP-Lactate Signaling Axis in Oligodendrocytes Regulates Whole-Body Metabolism. Cell Rep. 2019, 26, 2984–2997.e4. [Google Scholar] [CrossRef]
  57. Maekawa, A.; Balestrieri, B.; Austen, K.F.; Kanaoka, Y. GPR17 is a negative regulator of the cysteinyl leukotriene 1 receptor response to leukotriene D4. Proc. Natl. Acad. Sci. USA 2009, 106, 11685–11690. [Google Scholar] [CrossRef]
  58. Pugliese, A.M.; Trincavelli, M.L.; Lecca, D.; Coppi, E.; Fumagalli, M.; Ferrario, S.; Failli, P.; Daniele, S.; Martini, C.; Pedata, F.; et al. Functional characterization of two isoforms of the P2Y-like receptor GPR17: [35S]GTPγS binding and electrophysiological studies in 1321N1 cells. Am. J. Physiol. Cell Physiol. 2009, 297, C1028–C1040. [Google Scholar] [CrossRef]
  59. Hennen, S.; Wang, H.; Peters, L.; Merten, N.; Simon, K.; Spinrath, A.; Blättermann, S.; Akkari, R.; Schrage, R.; Schröder, R.; et al. Decoding signaling and function of the orphan G protein-coupled receptor GPR17 with a small-molecule agonist. Sci. Signal. 2013, 6, ra93. [Google Scholar] [CrossRef] [PubMed]
  60. 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. [Google Scholar] [CrossRef] [PubMed]
  61. Gelosa, P.; Colazzo, F.; Tremoli, E.; Sironi, L.; Castiglioni, L. Cysteinyl Leukotrienes as Potential Pharmacological Targets for Cerebral Diseases. Mediat. Inflamm. 2017, 2017, 3454212. [Google Scholar] [CrossRef] [PubMed]
  62. Zhao, C.Z.; Zhao, B.; Zhang, X.Y.; Huang, X.Q.; Shi, W.Z.; Liu, H.L.; Fang, S.H.; Lu, Y.B.; Zhang, W.P.; Tang, F.D.; 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. [Google Scholar] [CrossRef]
  63. Zhang, W.P.; Hu, H.; Zhang, L.; Ding, W.; Yao, H.T.; Chen, K.D.; Sheng, W.W.; Chen, Z.; Wei, E.Q. Expression of cysteinyl leukotriene receptor 1 in human traumatic brain injury and brain tumors. Neurosci. Lett. 2004, 363, 247–251. [Google Scholar] [CrossRef]
  64. Zhang, Y.-j.; Zhang, L.; Ye, Y.-l.; Fang, S.-h.; Zhou, Y.; Zhang, W.-p.; Lu, Y.-b.; Wei, E.-q. Cysteinyl leukotriene receptors CysLT1 and CysLT2 are upregulated in acute neuronal injury after focal cerebral ischemia in mice. Acta Pharmacol. Sin. 2006, 27, 1553–1560. [Google Scholar] [CrossRef]
  65. Fang, S.H.; Wei, E.Q.; Zhou, Y.; Wang, M.L.; Zhang, W.P.; Yu, G.L.; Chu, L.S.; Chen, Z. Increased expression of cysteinyl leukotriene receptor-1 in the brain mediates neuronal damage and astrogliosis after focal cerebral ischemia in rats. Neuroscience 2006, 140, 969–979. [Google Scholar] [CrossRef]
  66. Wang, Y.; Yang, Y.; Zhang, S.; Li, C.; Zhang, L. Modulation of neuroinflammation by cysteinyl leukotriene 1 and 2 receptors: Implications for cerebral ischemia and neurodegenerative diseases. Neurobiol. Aging 2020, 87, 1–10. [Google Scholar] [CrossRef]
  67. Michael, J.; Marschallinger, J.; Aigner, L. The leukotriene signaling pathway: A druggable target in Alzheimer’s disease. Drug Discov. Today 2019, 24, 505–516. [Google Scholar] [CrossRef] [PubMed]
  68. DiSabato, D.J.; Quan, N.; Godbout, J.P. Neuroinflammation: The devil is in the details. J. Neurochem. 2016, 139 (Suppl. S2), 136–153. [Google Scholar] [CrossRef] [PubMed]
  69. Shabab, T.; Khanabdali, R.; Moghadamtousi, S.Z.; Kadir, H.A.; Mohan, G. Neuroinflammation pathways: A general review. Int. J. Neurosci. 2017, 127, 624–633. [Google Scholar] [CrossRef]
  70. Skaper, S.D.; Facci, L.; Zusso, M.; Giusti, P. An Inflammation-Centric View of Neurological Disease: Beyond the Neuron. Front. Cell Neurosci. 2018, 12, 72. [Google Scholar] [CrossRef] [PubMed]
  71. Medzhitov, R. Origin and physiological roles of inflammation. Nature 2008, 454, 428–435. [Google Scholar] [CrossRef] [PubMed]
  72. Kawasaki, Y.; Zhang, L.; Cheng, J.K.; Ji, R.R. Cytokine mechanisms of central sensitization: Distinct and overlapping role of interleukin-1β, interleukin-6, and tumor necrosis factor-α in regulating synaptic and neuronal activity in the superficial spinal cord. J. Neurosci. 2008, 28, 5189–5194. [Google Scholar] [CrossRef] [PubMed]
  73. Schonberg, D.L.; Popovich, P.G.; McTigue, D.M. Oligodendrocyte generation is differentially influenced by toll-like receptor (TLR) 2 and TLR4-mediated intraspinal macrophage activation. J. Neuropathol. Exp. Neurol. 2007, 66, 1124–1135. [Google Scholar] [CrossRef]
  74. Sica, A.; Schioppa, T.; Mantovani, A.; Allavena, P. Tumour-associated macrophages are a distinct M2 polarised population promoting tumour progression: Potential targets of anti-cancer therapy. Eur. J. Cancer 2006, 42, 717–727. [Google Scholar] [CrossRef] [PubMed]
  75. Kotter, M.R.; Setzu, A.; Sim, F.J.; Van Rooijen, N.; Franklin, R.J. Macrophage depletion impairs oligodendrocyte remyelination following lysolecithin-induced demyelination. Glia 2001, 35, 204–212. [Google Scholar] [CrossRef]
  76. Tarr, A.J.; Liu, X.; Reed, N.S.; Quan, N. Kinetic characteristics of euflammation: The induction of controlled inflammation without overt sickness behavior. Brain Behav. Immun. 2014, 42, 96–108. [Google Scholar] [CrossRef] [PubMed]
  77. Liu, X.; Nemeth, D.P.; Tarr, A.J.; Belevych, N.; Syed, Z.W.; Wang, Y.; Ismail, A.S.; Reed, N.S.; Sheridan, J.F.; Yajnik, A.R.; et al. Euflammation attenuates peripheral inflammation-induced neuroinflammation and mitigates immune-to-brain signaling. Brain Behav. Immun. 2016, 54, 140–148. [Google Scholar] [CrossRef] [PubMed]
  78. Gyoneva, S.; Traynelis, S.F. Norepinephrine modulates the motility of resting and activated microglia via different adrenergic receptors. J. Biol. Chem. 2013, 288, 15291–15302. [Google Scholar] [CrossRef]
  79. Iwata, M.; Ota, K.T.; Duman, R.S. The inflammasome: Pathways linking psychological stress, depression, and systemic illnesses. Brain Behav. Immun. 2013, 31, 105–114. [Google Scholar] [CrossRef] [PubMed]
  80. Yoshida, H.; Goedert, M. Molecular cloning and functional characterization of chicken brain tau: Isoforms with up to five tandem repeats. Biochemistry-Us 2002, 41, 15203–15211. [Google Scholar] [CrossRef]
  81. Wohleb, E.S.; Powell, N.D.; Godbout, J.P.; Sheridan, J.F. Stress-induced recruitment of bone marrow-derived monocytes to the brain promotes anxiety-like behavior. J. Neurosci. 2013, 33, 13820–13833. [Google Scholar] [CrossRef]
  82. Mansour, R.M.; Ahmed, M.A.E.; El-Sahar, A.E.; El Sayed, N.S. Montelukast attenuates rotenone-induced microglial activation/p38 MAPK expression in rats: Possible role of its antioxidant, anti-inflammatory and antiapoptotic effects. Toxicol. Appl. Pharmacol. 2018, 358, 76–85. [Google Scholar] [CrossRef]
  83. Saad, M.A.; Abdelsalam, R.M.; Kenawy, S.A.; Attia, A.S. 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. [Google Scholar] [CrossRef]
  84. Tang, S.S.; Hong, H.; Chen, L.; Mei, Z.L.; Ji, M.J.; Xiang, G.Q.; Li, N.; Ji, H. Involvement of cysteinyl leukotriene receptor 1 in Aβ1-42-induced neurotoxicity in vitro and in vivo. Neurobiol. Aging 2014, 35, 590–599. [Google Scholar] [CrossRef]
  85. Hu, H.; Chen, G.; Zhang, J.M.; Zhang, W.P.; Zhang, L.; Ge, Q.F.; Yao, H.T.; Ding, W.; Chen, Z.; Wei, E.Q. Distribution of cysteinyl leukotriene receptor 2 in human traumatic brain injury and brain tumors. Acta Pharmacol Sin. 2005, 26, 685–690. [Google Scholar] [CrossRef] [PubMed]
  86. Chen, L.; Yang, Y.; Li, C.T.; Zhang, S.R.; Zheng, W.; Wei, E.Q.; Zhang, L.H. CysLT2 receptor mediates lipopolysaccharide-induced microglial inflammation and consequent neurotoxicity in vitro. Brain Res. 2015, 1624, 433–445. [Google Scholar] [CrossRef] [PubMed]
  87. Chen, F.; Ghosh, A.; Wu, F.; Tang, S.; Hu, M.; Sun, H.; Kong, L.; Hong, H. Preventive effect of genetic knockdown and pharmacological blockade of CysLT1R on lipopolysaccharide (LPS)-induced memory deficit and neurotoxicity in vivo. Brain Behav. Immun. 2017, 60, 255–269. [Google Scholar] [CrossRef]
  88. Shi, Q.J.; Wang, H.; Liu, Z.X.; Fang, S.H.; Song, X.M.; Lu, Y.B.; Zhang, W.P.; Sa, X.Y.; Ying, H.Z.; Wei, E.Q. 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. [Google Scholar] [CrossRef]
  89. Goodman & Gilman’s the Pharmacological Basis of Therapeutics, 3rd ed.; Brunton, L.L.; Knollmann, B.r.C.; Hilal-Dandan, R. (Eds.) McGraw Hill Medical: New York, NY, USA, 2018. [Google Scholar]
  90. Armada-Moreira, A.; Gomes, J.I.; Pina, C.C.; Savchak, O.K.; Gonçalves-Ribeiro, J.; Rei, N.; Pinto, S.; Morais, T.P.; Martins, R.S.; Ribeiro, F.F.; et al. Going the Extra (Synaptic) Mile: Excitotoxicity as the Road Toward Neurodegenerative Diseases. Front. Cell Neurosci. 2020, 14, 90. [Google Scholar] [CrossRef]
  91. Ding, Q.; Wei, E.Q.; Zhang, Y.J.; Zhang, W.P.; Chen, Z. Cysteinyl leukotriene receptor 1 is involved in N-methyl-D-aspartate-mediated neuronal injury in mice. Acta Pharmacol. Sin. 2006, 27, 1526–1536. [Google Scholar] [CrossRef]
  92. Kang, K.-H.; Liou, H.-H.; Hour, M.-J.; Liou, H.-C.; Fu, W.-M. Protection of dopaminergic neurons by 5-lipoxygenase inhibitor. Neuropharmacology 2013, 73, 380–387. [Google Scholar] [CrossRef] [PubMed]
  93. Raison, C.L.; Capuron, L.; Miller, A.H. Cytokines sing the blues: Inflammation and the pathogenesis of depression. Trends Immunol. 2006, 27, 24–31. [Google Scholar] [CrossRef]
  94. Sukoff Rizzo, S.J.; Neal, S.J.; Hughes, Z.A.; Beyna, M.; Rosenzweig-Lipson, S.; Moss, S.J.; Brandon, N.J. Evidence for sustained elevation of IL-6 in the CNS as a key contributor of depressive-like phenotypes. Transl. Psychiatry 2012, 2, e199. [Google Scholar] [CrossRef]
  95. Dantzer, R.; O’Connor, J.C.; Freund, G.G.; Johnson, R.W.; Kelley, K.W. From inflammation to sickness and depression: When the immune system subjugates the brain. Nat. Rev. Neurosci. 2008, 9, 46–56. [Google Scholar] [CrossRef] [PubMed]
  96. Bluthé, R.M.; Layé, S.; Michaud, B.; Combe, C.; Dantzer, R.; Parnet, P. Role of interleukin-1β and tumour necrosis factor-α in lipopolysaccharide-induced sickness behaviour: A study with interleukin-1 type I receptor-deficient mice. Eur. J. Neurosci. 2000, 12, 4447–4456. [Google Scholar] [CrossRef] [PubMed]
  97. Buschbeck, M.; Ghomashchi, F.; Gelb, M.H.; Watson, S.P.; Börsch-Haubold, A.G. Stress stimuli increase calcium-induced arachidonic acid release through phosphorylation of cytosolic phospholipase A2. Biochem. J. 1999, 344 Pt 2, 359–366. [Google Scholar] [CrossRef]
  98. Yu, X.B.; Dong, R.R.; Wang, H.; Lin, J.R.; An, Y.Q.; Du, Y.; Tang, S.S.; Hu, M.; Long, Y.; Sun, H.B.; 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. [Google Scholar] [CrossRef] [PubMed]
  99. Lin, J.R.; Fang, S.C.; Tang, S.S.; Hu, M.; Long, Y.; Ghosh, A.; Sun, H.B.; Kong, L.Y.; Hong, H. Hippocampal CysLT1R knockdown or blockade represses LPS-induced depressive behaviors and neuroinflammatory response in mice. Acta Pharmacol. Sin. 2017, 38, 477–487. [Google Scholar] [CrossRef]
  100. Uz, T.; Dimitrijevic, N.; Imbesi, M.; Manev, H.; Manev, R. Effects of MK-886, a 5-lipoxygenase activating protein (FLAP) inhibitor, and 5-lipoxygenase deficiency on the forced swimming behavior of mice. Neurosci. Lett. 2008, 436, 269–272. [Google Scholar] [CrossRef]
  101. Na, J.Y.; Song, K.; Lee, J.W.; Kim, S.; Kwon, J. 6-Shogaol has anti-amyloidogenic activity and ameliorates Alzheimer’s disease via CysLT1R-mediated inhibition of cathepsin B. Biochem. Biophys. Res. Commun. 2016, 477, 96–102. [Google Scholar] [CrossRef]
  102. Wang, X.Y.; Tang, S.S.; Hu, M.; Long, Y.; Li, Y.Q.; Liao, M.X.; Ji, H.; Hong, H. Leukotriene D4 induces amyloid-β generation via CysLT1R-mediated NF-κB pathways in primary neurons. Neurochem. Int. 2013, 62, 340–347. [Google Scholar] [CrossRef]
  103. Tang, S.S.; Wang, X.Y.; Hong, H.; Long, Y.; Li, Y.Q.; Xiang, G.Q.; Jiang, L.Y.; Zhang, H.T.; Liu, L.P.; Miao, M.X.; et al. Leukotriene D4 induces cognitive impairment through enhancement of CysLT1R-mediated amyloid-β generation in mice. Neuropharmacology 2013, 65, 182–192. [Google Scholar] [CrossRef]
  104. Herbst-Robinson, K.J.; Liu, L.; James, M.; Yao, Y.; Xie, S.X.; Brunden, K.R. Inflammatory Eicosanoids Increase Amyloid Precursor Protein Expression via Activation of Multiple Neuronal Receptors. Sci. Rep. 2015, 5, 18286. [Google Scholar] [CrossRef]
  105. Lai, J.; Hu, M.; Wang, H.; Hu, M.; Long, Y.; Miao, M.X.; Li, J.C.; Wang, X.B.; Kong, L.Y.; Hong, H. Montelukast targeting the cysteinyl leukotriene receptor 1 ameliorates Aβ1-42-induced memory impairment and neuroinflammatory and apoptotic responses in mice. Neuropharmacology 2014, 79, 707–714. [Google Scholar] [CrossRef] [PubMed]
  106. Kalra, J.; Kumar, P.; Majeed, A.B.; Prakash, A. Modulation of LOX and COX pathways via inhibition of amyloidogenesis contributes to mitoprotection against β-amyloid oligomer-induced toxicity in an animal model of Alzheimer's disease in rats. Pharmacol. Biochem. Behav. 2016, 146–147, 1–12. [Google Scholar] [CrossRef]
  107. Jang, H.; Kim, S.; Lee, J.M.; Oh, Y.-S.; Park, S.M.; Kim, S.R. Montelukast treatment protects nigral dopaminergic neurons against microglial activation in the 6-hydroxydopamine mouse model of Parkinson’s disease. Neuroreport 2017, 28, 242–249. [Google Scholar] [CrossRef] [PubMed]
  108. Bournival, J.; Plouffe, M.; Renaud, J.; Provencher, C.; Martinoli, M.G. Quercetin and sesamin protect dopaminergic cells from MPP+-induced neuroinflammation in a microglial (N9)-neuronal (PC12) coculture system. Oxid. Med. Cell Longev. 2012, 2012, 921941. [Google Scholar] [CrossRef] [PubMed]
  109. Chung, Y.C.; Kim, S.R.; Park, J.Y.; Chung, E.S.; Park, K.W.; Won, S.Y.; Bok, E.; Jin, M.; Park, E.S.; Yoon, S.H.; et al. Fluoxetine prevents MPTP-induced loss of dopaminergic neurons by inhibiting microglial activation. Neuropharmacology 2011, 60, 963–974. [Google Scholar] [CrossRef]
  110. Sherer, T.B.; Betarbet, R.; Kim, J.H.; Greenamyre, J.T. Selective microglial activation in the rat rotenone model of Parkinson’s disease. Neurosci. Lett. 2003, 341, 87–90. [Google Scholar] [CrossRef]
  111. Nagarajan, V.B.; Marathe, P.A. Effect of montelukast in experimental model of Parkinson’s disease. Neurosci. Lett. 2018, 682, 100–105. [Google Scholar] [CrossRef]
  112. GBD 2019 Diseases and Injuries Collaborators. Global burden of 369 diseases and injuries in 204 countries and territories, 1990–2019: A systematic analysis for the Global Burden of Disease Study 2019. Lancet 2020, 396, 1204–1222. [Google Scholar] [CrossRef]
  113. Reiss, T.F.; Altman, L.C.; Chervinsky, P.; Bewtra, A.; Stricker, W.E.; Noonan, G.P.; Kundu, S.; Zhang, J. Effects of montelukast (MK-0476), a new potent cysteinyl leukotriene (LTD4) receptor antagonist, in patients with chronic asthma. J. Allergy Clin. Immunol. 1996, 98, 528–534. [Google Scholar] [CrossRef]
  114. Knorr, B.; Matz, J.; Bernstein, J.A.; Nguyen, H.; Seidenberg, B.C.; Reiss, T.F.; Becker, A. Montelukast for chronic asthma in 6- to 14-year-old children: A randomized, double-blind trial. Pediatric Montelukast Study Group. JAMA 1998, 279, 1181–1186. [Google Scholar] [CrossRef]
  115. De Lepeleire, I.; Reiss, T.F.; Rochette, F.; Botto, A.; Zhang, J.; Kundu, S.; Decramer, M. Montelukast causes prolonged, potent leukotriene D4-receptor antagonism in the airways of patients with asthma. Clin. Pharmacol. Ther. 1997, 61, 83–92. [Google Scholar] [CrossRef]
  116. Araújo, A.C.; Tang, X.; Haeggström, J.Z. Targeting cysteinyl-leukotrienes in abdominal aortic aneurysm. Prostaglandins Other Lipid. Mediat. 2018, 139, 24–28. [Google Scholar] [CrossRef] [PubMed]
  117. Ramires, R.; Caiaffa, M.F.; Tursi, A.; Haeggström, J.Z.; Macchia, L. Novel inhibitory effect on 5-lipoxygenase activity by the anti-asthma drug montelukast. Biochem. Biophys. Res. Commun. 2004, 324, 815–821. [Google Scholar] [CrossRef]
  118. Trinh, H.K.T.; Suh, D.H.; Nguyen, T.V.T.; Choi, Y.; Park, H.S.; Shin, Y.S. Characterization of cysteinyl leukotriene-related receptors and their interactions in a mouse model of asthma. Prostaglandins Leukot. Essent. Fatty Acids 2019, 141, 17–23. [Google Scholar] [CrossRef]
  119. Goshtasbi, K.; Abouzari, M.; Abiri, A.; Ziai, K.; Lehrich, B.M.; Risbud, A.; Bayginejad, S.; Lin, H.W.; Djalilian, H.R. Trends and patterns of neurotology drug prescriptions on a nationwide insurance database. Laryngoscope Investig. Otolaryngol. 2021, 6, 1096–1103. [Google Scholar] [CrossRef] [PubMed]
  120. Balani, S.K.; Xu, X.; Pratha, V.; Koss, M.A.; Amin, R.D.; Dufresne, C.; Miller, R.R.; Arison, B.H.; Doss, G.A.; Chiba, M.; et al. Metabolic profiles of montelukast sodium (Singulair), a potent cysteinyl leukotriene1 receptor antagonist, in human plasma and bile. Drug Metab. Dispos. 1997, 25, 1282–1287. [Google Scholar]
  121. Chiba, M.; Xu, X.; Nishime, J.A.; Balani, S.K.; Lin, J.H. Hepatic microsomal metabolism of montelukast, a potent leukotriene D4 receptor antagonist, in humans. Drug Metab. Dispos. 1997, 25, 1022–1031. [Google Scholar] [PubMed]
  122. VandenBrink, B.M.; Foti, R.S.; Rock, D.A.; Wienkers, L.C.; Wahlstrom, J.L. Evaluation of CYP2C8 inhibition in vitro: Utility of montelukast as a selective CYP2C8 probe substrate. Drug Metab. Dispos. 2011, 39, 1546–1554. [Google Scholar] [CrossRef]
  123. Filppula, A.M.; Laitila, J.; Neuvonen, P.J.; Backman, J.T. Reevaluation of the microsomal metabolism of montelukast: Major contribution by CYP2C8 at clinically relevant concentrations. Drug Metab. Dispos. 2011, 39, 904–911. [Google Scholar] [CrossRef]
  124. Cardoso, J.O.; Oliveira, R.V.; Lu, J.B.; Desta, Z. In Vitro Metabolism of Montelukast by Cytochrome P450s and UDP-Glucuronosyltransferases. Drug Metab. Dispos. 2015, 43, 1905–1916. [Google Scholar] [CrossRef]
  125. Hirvensalo, P.; Tornio, A.; Neuvonen, M.; Tapaninen, T.; Paile-Hyvärinen, M.; Kärjä, V.; Männistö, V.T.; Pihlajamäki, J.; Backman, J.T.; Niemi, M. Comprehensive Pharmacogenomic Study Reveals an Important Role of UGT1A3 in Montelukast Pharmacokinetics. Clin. Pharmacol. Ther. 2018, 104, 158–168. [Google Scholar] [CrossRef] [PubMed]
  126. Cheng, H.; Leff, J.A.; Amin, R.; Gertz, B.J.; De Smet, M.; Noonan, N.; Rogers, J.D.; Malbecq, W.; Meisner, D.; Somers, G. Pharmacokinetics, bioavailability, and safety of montelukast sodium (MK-0476) in healthy males and females. Pharm. Res. 1996, 13, 445–448. [Google Scholar] [CrossRef] [PubMed]
  127. Merck Sharp & Dohme Corp. Full Prescribing Information: Singulair (Revised 6/2021). Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/020829s073,020830s075,021409s051lbl.pdf (accessed on 7 July 2018).
  128. Mougey, E.B.; Feng, H.; Castro, M.; Irvin, C.G.; Lima, J.J. Absorption of montelukast is transporter mediated: A common variant of OATP2B1 is associated with reduced plasma concentrations and poor response. Pharm. Genom. 2009, 19, 129–138. [Google Scholar] [CrossRef] [PubMed]
  129. Brännström, M.; Nordell, P.; Bonn, B.; Davis, A.M.; Palmgren, A.P.; Hilgendorf, C.; Rubin, K.; Grime, K. Montelukast Disposition: No Indication of Transporter-Mediated Uptake in OATP2B1 and OATP1B1 Expressing HEK293 Cells. Pharmaceutics 2015, 7, 554–564. [Google Scholar] [CrossRef] [PubMed]
  130. Bednarczyk, D.; Sanghvi, M.V. Organic anion transporting polypeptide 2B1 (OATP2B1), an expanded substrate profile, does it align with OATP2B1’s hypothesized function? Xenobiotica 2020, 50, 1128–1137. [Google Scholar] [CrossRef] [PubMed]
  131. Kinzi, J.; Grube, M.; Meyer Zu Schwabedissen, H.E. OATP2B1—The underrated member of the organic anion transporting polypeptide family of drug transporters? Biochem. Pharmacol. 2021, 188, 114534. [Google Scholar] [CrossRef] [PubMed]
  132. Clarridge, K.; Chin, S.; Eworuke, E.; Seymour, S. A Boxed Warning for Montelukast: The FDA Perspective. J. Allergy Clin. Immunol. Pract. 2021, 9, 2638–2641. [Google Scholar] [CrossRef]
  133. Calapai, G.; Casciaro, M.; Miroddi, M.; Calapai, F.; Navarra, M.; Gangemi, S. Montelukast-induced adverse drug reactions: A review of case reports in the literature. Pharmacology 2014, 94, 60–70. [Google Scholar] [CrossRef]
  134. Sansing-Foster, V.; Haug, N.; Mosholder, A.; Cocoros, N.M.; Bradley, M.; Ma, Y.; Pennap, D.; Dee, E.C.; Toh, S.; Pestine, E.; et al. Risk of Psychiatric Adverse Events Among Montelukast Users. J. Allergy Clin. Immunol. Pract. 2021, 9, 385–393.e12. [Google Scholar] [CrossRef]
  135. Uppsala Monitoring Centre. VigiAccess—WHO Programme for International Drug Monitoring. Available online: https://www.vigiaccess.org/ (accessed on 10 October 2021).
  136. Glockler-Lauf, S.D.; Finkelstein, Y.; Zhu, J.; Feldman, L.Y.; To, T. Montelukast and Neuropsychiatric Events in Children with Asthma: A Nested Case-Control Study. J. Pediatr. 2019, 209, 176–182.e4. [Google Scholar] [CrossRef]
  137. Aldea Perona, A.; García-Sáiz, M.; Sanz Álvarez, E. Psychiatric Disorders and Montelukast in Children: A Disproportionality Analysis of the VigiBase®. Drug Saf. 2016, 39, 69–78. [Google Scholar] [CrossRef] [PubMed]
  138. Kocyigit, A.; Gulcan Oksuz, B.; Yarar, F.; Uzun, F.; Igde, M.; Islek, I. Hallucination development with montelukast in a child with asthma: Case presentation. Iran. J. Allergy Asthma. Immunol. 2013, 12, 397–399. [Google Scholar] [PubMed]
  139. Byrne, F.; Oluwole, B.; Whyte, V.; Fahy, S.; McGuinness, D. Delayed Onset of Neuropsychiatric Effects Associated with Montelukast. Ir. J. Psychol. Med. 2012, 29, 125–127. [Google Scholar] [CrossRef] [PubMed]
  140. Bernal Vañó, E.; López Andrés, N. Un caso de síndrome de Alicia en el país de las maravillas en probable relación con el uso de montelukast [A case of Alice-in-Wonderland syndrome probably associated with the use of montelukast]. An. Pediatr. (Barc) 2013, 78, 127–128. [Google Scholar] [CrossRef]
  141. Carnovale, C.; Gentili, M.; Antoniazzi, S.; Radice, S.; Clementi, E. Montelukast-induced metamorphopsia in a pediatric patient: A case report and a pharmacovigilance database analysis. Ann. Allergy Asthma. Immunol. 2016, 116, 370–371. [Google Scholar] [CrossRef]
  142. Benard, B.; Bastien, V.; Vinet, B.; Yang, R.; Krajinovic, M.; Ducharme, F.M. Neuropsychiatric adverse drug reactions in children initiated on montelukast in real-life practice. Eur. Respir. J. 2017, 50, 1700148. [Google Scholar] [CrossRef]
  143. Wallerstedt, S.M.; Brunlöf, G.; Sundström, A.; Eriksson, A.L. Montelukast and psychiatric disorders in children. Pharmacoepidemiol. Drug Saf. 2009, 18, 858–864. [Google Scholar] [CrossRef]
  144. Caudevilla Lafuente, P.; Garcia Íñiguez, J.P.; Martín de Vicente, C. Reacciones adversas a montelukast: De la teoría a la práctica. Serie de casos [Adverse drug reactions of montelukast: From theory to practice. Case report]. Arch. Argent. Pediatr. 2021, 119, e357–e359. [Google Scholar] [CrossRef]
  145. Alkhuja, S.; Gazizov, N.; Alexander, M.E. Sleeptalking! Sleepwalking! Side effects of montelukast. Case Rep. Pulmonol. 2013, 2013, 813786. [Google Scholar] [CrossRef]
  146. Anandan, N.; Ibitoye, F. Montelukast and worsening of hallucinations in paranoid schizophrenia. Psychiatr. Bulletin. 2008, 32, 276. [Google Scholar] [CrossRef]
  147. Ibarra-Barrueta, O.; Palacios-Zabalza, I.; Mora-Atorrasagasti, O.; Mayo-Suarez, J. Effect of concomitant use of montelukast and efavirenz on neuropsychiatric adverse events. Ann. Pharm. 2014, 48, 145–148. [Google Scholar] [CrossRef]
  148. Philip, G.; Hustad, C.; Noonan, G.; Malice, M.-P.; Ezekowitz, A.; Reiss, T.F.; Knorr, B. Reports of suicidality in clinical trials of montelukast. J. Allergy Clin. Immunol. 2009, 124, 691–696.e6. [Google Scholar] [CrossRef]
  149. Jick, H.; Hagberg, K.W.; Egger, P. Rate of suicide in patients taking montelukast. Pharmacotherapy 2009, 29, 165–166. [Google Scholar] [CrossRef] [PubMed]
  150. Manalai, P.; Woo, J.-M.; Postolache, T.T. Suicidality and montelukast. Expert Opin. Drug Saf. 2009, 8, 273–282. [Google Scholar] [CrossRef]
  151. Schumock, G.T.; Stayner, L.T.; Valuck, R.J.; Joo, M.J.; Gibbons, R.D.; Lee, T.A. Risk of suicide attempt in asthmatic children and young adults prescribed leukotriene-modifying agents: A nested case-control study. J. Allergy Clin. Immunol. 2012, 130, 368–375. [Google Scholar] [CrossRef] [PubMed]
  152. Das, S.; Mondal, S.; Dey, J.K.; Bandyopadhyay, S.; Saha, I.; Tripathi, S.K. A case of montelukast induced hypercholesterolemia, severe hypertriglyceridemia and pancreatitis. J. Young Pharm. 2013, 5, 64–66. [Google Scholar] [CrossRef]
  153. Xie, J.-X.; Wei, J.-F.; Meng, L. Montelukast sodium-induced hematuria: A case report and literature review of 19 cases in mainland China. Int J. Clin. Pharm. Ther. 2013, 51, 958–962. [Google Scholar] [CrossRef]
  154. Harugeri, A.; Parthasarathi, G.; Sharma, J.; D’Souza, G.A.; Ramesh, M. Montelukast induced acute hepatocellular liver injury. J. Postgrad. Med. 2009, 55, 141–142. [Google Scholar] [CrossRef]
  155. Russmann, S.; Iselin, H.U.; Meier, D.; Zimmermann, A.; Simon, H.-U.; Caduff, P.; Reichen, J. Acute hepatitis associated with montelukast. J. Hepatol. 2003, 38, 694–695. [Google Scholar] [CrossRef]
  156. Incecik, F.; Onlen, Y.; Sangun, O.; Akoglu, S. Probable montelukast-induced hepatotoxicity in a pediatric patient: Case report. Ann. Saudi. Med. 2007, 27, 462–463. [Google Scholar] [CrossRef] [PubMed]
  157. Lebensztejn, D.M.; Bobrus-Chociej, A.; Kłusek, M.; Uscinowicz, M.; Lotowska, J.; Sobaniec-Lotowska, M.; Kaczmarski, M. Hepatotoxicity caused by montelukast in a paediatric patient. Prz. Gastroenterol. 2014, 9, 121–123. [Google Scholar] [CrossRef]
  158. Sabbagh, R.; Sheikh-Taha, M. Possible montelukast-induced angioedema. Am. J. Health Syst. Pharm. 2009, 66, 1705–1706. [Google Scholar] [CrossRef]
  159. Minciullo, P.L.; Saija, A.; Bonanno, D.; Ferlazzo, E.; Gangemi, S. Montelukast-induced generalized urticaria. Ann. Pharm. 2004, 38, 999–1001. [Google Scholar] [CrossRef] [PubMed]
  160. Cetkovská, P.; Pizinger, K. Childhood pemphigus associated with montelukast administration. Clin. Exp. Dermatol. 2003, 28, 328–329. [Google Scholar] [CrossRef] [PubMed]
  161. Price, D. Tolerability of montelukast. Drugs 2000, 59 (Suppl. S1), 35–42; discussion 43–45. [Google Scholar] [CrossRef] [PubMed]
  162. Haarman, M.G.; van Hunsel, F.; de Vries, T.W. Adverse drug reactions of montelukast in children and adults. Pharm. Res. Perspect. 2017, 5, e00341. [Google Scholar] [CrossRef]
  163. Hauser, T.; Mahr, A.; Metzler, C.; Coste, J.; Sommerstein, R.; Gross, W.L.; Guillevin, L.; Hellmich, B. The leucotriene receptor antagonist montelukast and the risk of Churg-Strauss syndrome: A case-crossover study. Thorax 2008, 63, 677–682. [Google Scholar] [CrossRef] [PubMed]
  164. Anar, C.; Ünsal, I.; Ozanturk, M.E.; Halilçolar, H.; Yucel, N. A case of Churg-Strauss syndrome treated with montelukast. Med. Princ. Pract. 2012, 21, 186–189. [Google Scholar] [CrossRef]
  165. Mateo, M.L.; Cortés, C.M.; Berisa, F. Síndrome de Churg-Strauss asociado a la administración de montelukast en un paciente asmático sin tratamiento esteroide de base. Arch. Bronconeumol. 2002, 38, 56. [Google Scholar] [CrossRef]
  166. Jennings, L.; Ho, W.L.; Gulmann, C.; Murphy, G.M. Churg-Strauss syndrome secondary to antileucotriene therapy in a patient not receiving oral corticosteroids. Clin. Exp. Dermatol. 2009, 34, e430–e431. [Google Scholar] [CrossRef]
  167. Churg, J.; Strauss, L. Allergic granulomatosis, allergic angiitis, and periarteritis nodosa. Am. J. Pathol 1951, 27, 277–301. [Google Scholar]
  168. Franco, J.; Artés, M.J. Pulmonary eosinophilia associated with montelukast. Thorax 1999, 54, 558–560. [Google Scholar] [CrossRef]
  169. Wechsler, M.E.; Finn, D.; Gunawardena, D.; Westlake, R.; Barker, A.; Haranath, S.P.; Pauwels, R.A.; Kips, J.C.; Drazen, J.M. Churg-Strauss syndrome in patients receiving montelukast as treatment for asthma. Chest 2000, 117, 708–713. [Google Scholar] [CrossRef]
  170. Villena, V.; Hidalgo, R.; Sotelo, M.T.; Martin-Escribano, P. Montelukast and Churg-Strauss syndrome. Eur. Respir. J. 2000, 15, 626. [Google Scholar] [CrossRef] [PubMed]
  171. Uyar, M.; Elbek, O.; Bakır, K.; Kibar, Y.; Bayram, N.; Dikensoy, Ö. Churg-Strauss syndrome related to montelukast. Tuberk. Toraks 2012, 60, 56–58. [Google Scholar] [CrossRef] [PubMed]
  172. Tang, M.B.; Yosipovitch, G. Acute Churg-Strauss syndrome in an asthmatic patient receiving montelukast therapy. Arch. Dermatol. 2003, 139, 715–718. [Google Scholar] [CrossRef]
  173. Cuchacovich, R.; Justiniano, M.; Espinoza, L.R. Churg-Strauss syndrome associated with leukotriene receptor antagonists (LTRA). Clin. Rheumatol. 2007, 26, 1769–1771. [Google Scholar] [CrossRef]
  174. Kaliterna, D.M.; Perković, D.; Radić, M. Churg-Strauss syndrome associated with montelukast therapy. J. Asthma. 2009, 46, 604–605. [Google Scholar] [CrossRef] [PubMed]
  175. Black, J.G.; Bonner, J.R.; Boulware, D.; Andea, A.A. Montelukast-associated Churg-Strauss vasculitis: Another associated report. Ann. Allergy Asthma. Immunol. 2009, 102, 351–352. [Google Scholar] [CrossRef]
  176. Kanda, T.; Tanio, H.; Wu, C.; Nishihara, H.; Nogaki, F.; Ono, T. Churg-Strauss syndrome with severe granulomatous angiitis and crescentic glomerulonephritis, which developed during therapy with a leukotriene receptor antagonist. Clin. Exp. Nephrol. 2010, 14, 602–607. [Google Scholar] [CrossRef]
  177. Camozzi, P.; Milani, G.P.; Bianchetti, M.G. Leukotriene receptor antagonists in Henoch-Schonlein syndrome: Friends or foes? Pediatr. Int. 2014, 56, 802. [Google Scholar] [CrossRef] [PubMed]
  178. Rejnö, G.; Lundholm, C.; Gong, T.; Larsson, K.; Saltvedt, S.; Almqvist, C. Asthma during pregnancy in a population-based study--pregnancy complications and adverse perinatal outcomes. PLoS ONE 2014, 9, e104755. [Google Scholar] [CrossRef]
  179. Nelsen, L.M.; Shields, K.E.; Cunningham, M.L.; Stoler, J.M.; Bamshad, M.J.; Eng, P.M.; Smugar, S.S.; Gould, A.L.; Philip, G. Congenital malformations among infants born to women receiving montelukast, inhaled corticosteroids, and other asthma medications. J. Allergy Clin. Immunol. 2012, 129, 251–254.e6. [Google Scholar] [CrossRef] [PubMed]
  180. Sarkar, M.; Koren, G.; Kalra, S.; Ying, A.; Smorlesi, C.; De Santis, M.; Diav-Citrin, O.; Avgil, M.; Lavigne, S.V.; Berkovich, M.; et al. Montelukast use during pregnancy: A multicentre, prospective, comparative study of infant outcomes. Eur. J. Clin. Pharmacol. 2009, 65, 1259–1264. [Google Scholar] [CrossRef]
  181. Bakhireva, L.N.; Jones, K.L.; Schatz, M.; Klonoff-Cohen, H.S.; Johnson, D.; Slymen, D.J.; Chambers, C.D.; Organization of Teratology Information Specialists Collaborative Research Group. Safety of leukotriene receptor antagonists in pregnancy. J. Allergy Clin. Immunol. 2007, 119, 618–625. [Google Scholar] [CrossRef] [PubMed]
  182. Cavero-Carbonell, C.; Vinkel-Hansen, A.; Rabanque-Hernández, M.J.; Martos, C.; Garne, E. Fetal Exposure to Montelukast and Congenital Anomalies: A Population Based Study in Denmark. Birth Defects Res. 2017, 109, 452–459. [Google Scholar] [CrossRef]
  183. Lai, J.; Mei, Z.L.; Wang, H.; Hu, M.; Long, Y.; Miao, M.X.; Li, N.; Hong, H. Montelukast rescues primary neurons against Aβ1–42-induced toxicity through inhibiting CysLT1R-mediated NF-κB signaling. Neurochem. Int. 2014, 75, 26–31. [Google Scholar] [CrossRef]
  184. Zhang, X.Y.; Chen, L.; Yang, Y.; Xu, D.M.; Zhang, S.R.; Li, C.T.; Zheng, W.; Yu, S.Y.; Wei, E.Q.; Zhang, L.H. Regulation of rotenone-induced microglial activation by 5-lipoxygenase and cysteinyl leukotriene receptor 1. Brain Res. 2014, 1572, 59–71. [Google Scholar] [CrossRef]
  185. Wallin, J.; Svenningsson, P. Potential Effects of Leukotriene Receptor Antagonist Montelukast in Treatment of Neuroinflammation in Parkinson’s Disease. Int. J. Mol. Sci. 2021, 22, 5606. [Google Scholar] [CrossRef]
  186. Kalonia, H.; Kumar, P.; Kumar, A.; Nehru, B. Protective effect of montelukast against quinolinic acid/malonic acid induced neurotoxicity: Possible behavioral, biochemical, mitochondrial and tumor necrosis factor-α level alterations in rats. Neuroscience 2010, 171, 284–299. [Google Scholar] [CrossRef]
  187. Marschallinger, J.; Altendorfer, B.; Rockenstein, E.; Holztrattner, M.; Garnweidner-Raith, J.; Pillichshammer, N.; Leister, I.; Hutter-Paier, B.; Strempfl, K.; Unger, M.S.; et al. The Leukotriene Receptor Antagonist Montelukast Reduces Alpha-Synuclein Load and Restores Memory in an Animal Model of Dementia with Lewy Bodies. Neurotherapeutics 2020, 17, 1061–1074. [Google Scholar] [CrossRef]
  188. Grinde, B.; Schirmer, H.; Eggen, A.E.; Aigner, L.; Engdahl, B. A possible effect of montelukast on neurological aging examined by the use of register data. Int. J. Clin. Pharm. 2021, 43, 541–548. [Google Scholar] [CrossRef] [PubMed]
  189. Michael, J.; Zirknitzer, J.; Unger, M.S.; Poupardin, R.; Rieß, T.; Paiement, N.; Zerbe, H.; Hutter-Paier, B.; Reitsamer, H.; Aigner, L. The Leukotriene Receptor Antagonist Montelukast Attenuates Neuroinflammation and Affects Cognition in Transgenic 5xFAD Mice. Int. J. Mol. Sci. 2021, 22, 2782. [Google Scholar] [CrossRef] [PubMed]
  190. Zhang, C.T.; Lin, J.R.; Wu, F.; Ghosh, A.; Tang, S.S.; Hu, M.; Long, Y.; Sun, H.B.; Hong, H. Montelukast ameliorates streptozotocin-induced cognitive impairment and neurotoxicity in mice. Neurotoxicology 2016, 57, 214–222. [Google Scholar] [CrossRef] [PubMed]
  191. Tsai, M.J.; Chang, W.A.; Tsai, P.H.; Wu, C.Y.; Ho, Y.W.; Yen, M.C.; Lin, Y.S.; Kuo, P.L.; Hsu, Y.L. Montelukast Induces Apoptosis-Inducing Factor-Mediated Cell Death of Lung Cancer Cells. Int. J. Mol. Sci. 2017, 18, 1353. [Google Scholar] [CrossRef]
  192. Tsai, M.J.; Wu, P.H.; Sheu, C.C.; Hsu, Y.L.; Chang, W.A.; Hung, J.Y.; Yang, C.J.; Yang, Y.H.; Kuo, P.L.; Huang, M.S. Cysteinyl Leukotriene Receptor Antagonists Decrease Cancer Risk in Asthma Patients. Sci. Rep. 2016, 6, 23979. [Google Scholar] [CrossRef]
  193. Tang, C.; Lei, H.; Zhang, J.; Liu, M.; Jin, J.; Luo, H.; Xu, H.; Wu, Y. Montelukast inhibits hypoxia inducible factor-1α translation in prostate cancer cells. Cancer Biol. Ther. 2018, 19, 715–721. [Google Scholar] [CrossRef]
  194. Matsuyama, M.; Hayama, T.; Funao, K.; Kawahito, Y.; Sano, H.; Takemoto, Y.; Nakatani, T.; Yoshimura, R. Overexpression of cysteinyl LT1 receptor in prostate cancer and CysLT1R antagonist inhibits prostate cancer cell growth through apoptosis. Oncol. Rep. 2007, 18, 99–104. [Google Scholar] [CrossRef]
  195. Tahan, F.; Jazrawi, E.; Moodley, T.; Rovati, G.E.; Adcock, I.M. Montelukast inhibits tumour necrosis factor-alpha-mediated interleukin-8 expression through inhibition of nuclear factor-κB p65-associated histone acetyltransferase activity. Clin. Exp. Allergy 2008, 38, 805–811. [Google Scholar] [CrossRef]
  196. Sanghai, N.; Tranmer, G.K. Taming the cytokine storm: Repurposing montelukast for the attenuation and prophylaxis of severe COVID-19 symptoms. Drug Discov. Today 2020, 25, 2076–2079. [Google Scholar] [CrossRef]
  197. Funk, C.D.; Ardakani, A. A Novel Strategy to Mitigate the Hyperinflammatory Response to COVID-19 by Targeting Leukotrienes. Front. Pharmacol. 2020, 11, 1214. [Google Scholar] [CrossRef]
  198. Kow, C.S.; Hasan, S.S. Montelukast in children with allergic rhinitis amid COVID-19 pandemic. Acta Paediatr. 2020, 109, 2151. [Google Scholar] [CrossRef]
  199. Dey, M.; Singh, R.K. Possible Therapeutic Potential of Cysteinyl Leukotriene Receptor Antagonist Montelukast in Treatment of SARS-CoV-2-Induced COVID-19. Pharmacology 2021, 106, 469–476. [Google Scholar] [CrossRef]
  200. Aigner, L.; Pietrantonio, F.; Bessa de Sousa, D.M.; Michael, J.; Schuster, D.; Reitsamer, H.A.; Zerbe, H.; Studnicka, M. The Leukotriene Receptor Antagonist Montelukast as a Potential COVID-19 Therapeutic. Front. Mol. Biosci. 2020, 7, 610132. [Google Scholar] [CrossRef]
  201. Wixted, J.J.; Fanning, P.J.; Gaur, T.; O'Connell, S.L.; Silva, J.; Mason-Savas, A.; Ayers, D.C.; Stein, G.S.; Lian, J.B. Enhanced fracture repair by leukotriene antagonism is characterized by increased chondrocyte proliferation and early bone formation: A novel role of the cysteinyl LT-1 receptor. J. Cell Physiol. 2009, 221, 31–39. [Google Scholar] [CrossRef]
  202. Li, Z.; Wang, J.; Ma, Y. Montelukast attenuates interleukin IL-1β-induced oxidative stress and apoptosis in chondrocytes by inhibiting CYSLTR1 (Cysteinyl Leukotriene Receptor 1) and activating KLF2 (Kruppel Like Factor 2). Bioengineered 2021, 12, 8476–8484. [Google Scholar] [CrossRef] [PubMed]
  203. Ingelsson, E.; Yin, L.; Bäck, M. Nationwide cohort study of the leukotriene receptor antagonist montelukast and incident or recurrent cardiovascular disease. J. Allergy Clin. Immunol. 2012, 129, 702–707.e2. [Google Scholar] [CrossRef]
  204. Bäck, M.; Yin, L.; Nagy, E.; Ingelsson, E. The leukotriene receptor antagonist montelukast and aortic stenosis. Br. J. Clin. Pharmacol. 2013, 75, 280–281. [Google Scholar] [CrossRef]
  205. Allayee, H.; Hartiala, J.; Lee, W.; Mehrabian, M.; Irvin, C.G.; Conti, D.V.; Lima, J.J. The effect of montelukast and low-dose theophylline on cardiovascular disease risk factors in asthmatics. Chest 2007, 132, 868–874. [Google Scholar] [CrossRef] [PubMed]
  206. Alizamani, E.; Ghorbanzadeh, B.; Naserzadeh, R.; Mansouri, M.T. Montelukast, a cysteinyl leukotriene receptor antagonist, exerts local antinociception in animal model of pain through the L-arginine/nitric oxide/cyclic GMP/KATP channel pathway and PPARgamma receptors. Int. J. Neurosci. 2021, 131, 1004–1011. [Google Scholar] [CrossRef] [PubMed]
  207. Zhao, R.; Shi, W.-Z.; Zhang, Y.-M.; Fang, S.-H.; Wei, E.-Q. 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. [Google Scholar] [CrossRef] [PubMed]
  208. Lenz, Q.F.; Arroyo, D.S.; Temp, F.R.; Poersch, A.B.; Masson, C.J.; Jesse, A.C.; Marafiga, J.R.; Reschke, C.R.; Iribarren, P.; Mello, C.F. Cysteinyl leukotriene receptor (CysLT) antagonists decrease pentylenetetrazol-induced seizures and blood-brain barrier dysfunction. Neuroscience 2014, 277, 859–871. [Google Scholar] [CrossRef] [PubMed]
  209. Rehni, A.K.; Singh, T.G. Modulation of leukotriene D4 attenuates the development of seizures in mice. Prostaglandins Leukot. Essent. Fatty Acids 2011, 85, 97–106. [Google Scholar] [CrossRef] [PubMed]
  210. Wang, L.; Du, C.; Lv, J.; Wei, W.; Cui, Y.; Xie, X. 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. [Google Scholar] [CrossRef] [PubMed]
  211. Wu, C.; Liu, Y.; Yang, Y.; Zhang, P.; Zhong, W.; Wang, Y.; Wang, Q.; Xu, Y.; Li, M.; Li, X.; et al. Analysis of therapeutic targets for SARS-CoV-2 and discovery of potential drugs by computational methods. Acta Pharm. Sin. B 2020, 10, 766–788. [Google Scholar] [CrossRef] [PubMed]
  212. Copertino, D.C.; Duarte, R.R.R.; Powell, T.R.; de Mulder Rougvie, M.; Nixon, D.F. Montelukast drug activity and potential against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). J. Med. Virol. 2021, 93, 187–189. [Google Scholar] [CrossRef] [PubMed]
  213. Khan, A.R.; Misdary, C.; Yegya-Raman, N.; Kim, S.; Narayanan, N.; Siddiqui, S.; Salgame, P.; Radbel, J.; Groote, F.; Michel, C.; et al. Montelukast in hospitalized patients diagnosed with COVID-19. J. Asthma. 2021, 59, 1–7. [Google Scholar] [CrossRef]
  214. Tr Trost, A.; Motloch, K.; Koller, A.; Bruckner, D.; Runge, C.; Schroedl, F.; Bogner, B.; Kaser-Eichberger, A.; Strohmaier, C.; Ladek, A.M.; et al. Inhibition of the cysteinyl leukotriene pathways increases survival of RGCs and reduces microglial activation in ocular hypertension. Exp. Eye Res. 2021, 213, 108806. [Google Scholar] [CrossRef]
  215. Verleden, G.M.; Verleden, S.E.; Vos, R.; De Vleeschauwer, S.I.; Dupont, L.J.; Van Raemdonck, D.E.; Vanaudenaerde, B.M. Montelukast for bronchiolitis obliterans syndrome after lung transplantation: A pilot study. Transpl. Int. 2011, 24, 651–656. [Google Scholar] [CrossRef]
  216. Ruttens, D.; Verleden, S.E.; Demeyer, H.; Van Raemdonck, D.E.; Yserbyt, J.; Dupont, L.J.; Vanaudenaerde, B.M.; Vos, R.; Verleden, G.M. Montelukast for bronchiolitis obliterans syndrome after lung transplantation: A randomized controlled trial. PLoS ONE 2018, 13, e0193564. [Google Scholar] [CrossRef]
  217. Vos, R.; Eynde, R.V.; Ruttens, D.; Verleden, S.E.; Vanaudenaerde, B.M.; Dupont, L.J.; Yserbyt, J.; Verbeken, E.K.; Neyrinck, A.P.; Van Raemdonck, D.E.; et al. Montelukast in chronic lung allograft dysfunction after lung transplantation. J. Heart Lung Transplant. 2019, 38, 516–527. [Google Scholar] [CrossRef] [PubMed]
  218. Shimbori, C.; Shiota, N.; Okunishi, H. Effects of montelukast, a cysteinyl-leukotriene type 1 receptor antagonist, on the pathogenesis of bleomycin-induced pulmonary fibrosis in mice. Eur. J. Pharmacol. 2011, 650, 424–430. [Google Scholar] [CrossRef] [PubMed]
  219. Helmy, M.M.; El-Gowelli, H.M. Montelukast abrogates rhabdomyolysis-induced acute renal failure via rectifying detrimental changes in antioxidant profile and systemic cytokines and apoptotic factors production. Eur. J. Pharmacol. 2012, 683, 294–300. [Google Scholar] [CrossRef] [PubMed]
  220. Suddek, G.M. Montelukast ameliorates kidney function and urinary bladder sensitivity in experimentally induced renal dysfunction in rats. Fundam. Clin. Pharmacol. 2013, 27, 186–191. [Google Scholar] [CrossRef] [PubMed]
  221. Tuğtepe, H.; Şener, G.; Çetinel, S.; Velioğlu-Öğünç, A.; Yeğen, B.C. Oxidative renal damage in pyelonephritic rats is ameliorated by montelukast, a selective leukotriene CysLT1 receptor antagonist. Eur. J. Pharmacol. 2007, 557, 69–75. [Google Scholar] [CrossRef]
  222. Kort, E.; Jovinge, S. Drug Repurposing: Claiming the Full Benefit from Drug Development. Curr. Cardiol. Rep. 2021, 23, 62. [Google Scholar] [CrossRef]
  223. Dugger, B.N.; Dickson, D.W. Pathology of Neurodegenerative Diseases. Cold Spring Harb. Perspect. Biol. 2017, 9, a028035. [Google Scholar] [CrossRef]
  224. Wyss-Coray, T. Ageing, neurodegeneration and brain rejuvenation. Nature 2016, 539, 180–186. [Google Scholar] [CrossRef]
  225. Elobeid, A.; Libard, S.; Leino, M.; Popova, S.N.; Alafuzoff, I. Altered Proteins in the Aging Brain. J. Neuropathol. Exp. Neurol. 2016, 75, 316–325. [Google Scholar] [CrossRef]
  226. Müller, U.C.; Deller, T.; Korte, M. Not just amyloid: Physiological functions of the amyloid precursor protein family. Nat. Rev. Neurosci. 2017, 18, 281–298. [Google Scholar] [CrossRef]
  227. Tiwari, S.; Atluri, V.; Kaushik, A.; Yndart, A.; Nair, M. Alzheimer’s disease: Pathogenesis, diagnostics, and therapeutics. Int. J. Nanomed. 2019, 14, 5541–5554. [Google Scholar] [CrossRef] [PubMed]
  228. Alzheimer’s Association. 2020 Alzheimer’s disease facts and figures. Alzheimer’s Dement. 2020, 16, 391–460. [Google Scholar] [CrossRef]
  229. Naj, A.C.; Jun, G.; Beecham, G.W.; Wang, L.S.; Vardarajan, B.N.; Buros, J.; Gallins, P.J.; Buxbaum, J.D.; Jarvik, G.P.; Crane, P.K.; et al. Common variants at MS4A4/MS4A6E, CD2AP, CD33 and EPHA1 are associated with late-onset Alzheimer’s disease. Nat. Genet. 2011, 43, 436–441. [Google Scholar] [CrossRef]
  230. L Lambert, J.C.; Heath, S.; Even, G.; Campion, D.; Sleegers, K.; Hiltunen, M.; Combarros, O.; Zelenika, D.; Bullido, M.J.; Tavernier, B.; et al. Genome-wide association study identifies variants at CLU and CR1 associated with Alzheimer’s disease. Nat. Genet. 2009, 41, 1094–1099. [Google Scholar] [CrossRef] [PubMed]
  231. Hollingworth, P.; Harold, D.; Sims, R.; Gerrish, A.; Lambert, J.C.; Carrasquillo, M.M.; Abraham, R.; Hamshere, M.L.; Pahwa, J.S.; Moskvina, V.; et al. Common variants at ABCA7, MS4A6A/MS4A4E, EPHA1, CD33 and CD2AP are associated with Alzheimer’s disease. Nat. Genet. 2011, 43, 429–435. [Google Scholar] [CrossRef]
  232. Harold, D.; Abraham, R.; Hollingworth, P.; Sims, R.; Gerrish, A.; Hamshere, M.L.; Pahwa, J.S.; Moskvina, V.; Dowzell, K.; Williams, A.; et al. Genome-wide association study identifies variants at CLU and PICALM associated with Alzheimer’s disease. Nat. Genet. 2009, 41, 1088–1093. [Google Scholar] [CrossRef]
  233. Bellenguez, C.; Charbonnier, C.; Grenier-Boley, B.; Quenez, O.; Le Guennec, K.; Nicolas, G.; Chauhan, G.; Wallon, D.; Rousseau, S.; Richard, A.C.; et al. Contribution to Alzheimer’s disease risk of rare variants in TREM2, SORL1, and ABCA7 in 1779 cases and 1273 controls. Neurobiol. Aging 2017, 59, 220.e1–220.e9. [Google Scholar] [CrossRef] [PubMed]
  234. Mandelkow, E.M.; Mandelkow, E. Biochemistry and cell biology of tau protein in neurofibrillary degeneration. Cold Spring Harb. Perspect. Med. 2012, 2, a006247. [Google Scholar] [CrossRef]
  235. Viswanathan, A.; Greenberg, S.M. Cerebral amyloid angiopathy in the elderly. Ann. Neurol. 2011, 70, 871–880. [Google Scholar] [CrossRef]
  236. Querfurth, H.W.; LaFerla, F.M. Alzheimer’s disease. N. Engl. J. Med. 2010, 362, 329–344. [Google Scholar] [CrossRef]
  237. Robakis, N.K.; Wisniewski, H.M.; Jenkins, E.C.; Devine-Gage, E.A.; Houck, G.E.; Yao, X.L.; Ramakrishna, N.; Wolfe, G.; Silverman, W.P.; Brown, W.T. Chromosome 21q21 sublocalisation of gene encoding beta-amyloid peptide in cerebral vessels and neuritic (senile) plaques of people with Alzheimer disease and Down syndrome. Lancet 1987, 1, 384–385. [Google Scholar] [CrossRef]
  238. Carrodeguas, J.A.; Rodolosse, A.; Garza, M.V.; Sanz-Clemente, A.; Peréz-Pé, R.; Lacosta, A.M.; Domínguez, L.; Monleon, I.; Sánchez-Díaz, R.; Sorribas, V.; et al. The chick embryo appears as a natural model for research in beta-amyloid precursor protein processing. Neuroscience 2005, 134, 1285–1300. [Google Scholar] [CrossRef] [PubMed]
  239. De Strooper, B.; Vassar, R.; Golde, T. The secretases: Enzymes with therapeutic potential in Alzheimer disease. Nat. Rev. Neurol. 2010, 6, 99–107. [Google Scholar] [CrossRef] [PubMed]
  240. Chen, G.F.; Xu, T.H.; Yan, Y.; Zhou, Y.R.; Jiang, Y.; Melcher, K.; Xu, H.E. Amyloid beta: Structure, biology and structure-based therapeutic development. Acta Pharmacol. Sin. 2017, 38, 1205–1235. [Google Scholar] [CrossRef] [PubMed]
  241. Fu, L.; Sun, Y.; Guo, Y.; Chen, Y.; Yu, B.; Zhang, H.; Wu, J.; Yu, X.; Kong, W.; Wu, H. Comparison of neurotoxicity of different aggregated forms of Aβ40, Aβ42 and Aβ43 in cell cultures. J. Pept. Sci. 2017, 23, 245–251. [Google Scholar] [CrossRef]
  242. Blennow, K.; de Leon, M.J.; Zetterberg, H. Alzheimer’s disease. Lancet 2006, 368, 387–403. [Google Scholar] [CrossRef]
  243. Nalivaeva, N.N.; Turner, A.J. Targeting amyloid clearance in Alzheimer’s disease as a therapeutic strategy. Br. J. Pharmacol. 2019, 176, 3447–3463. [Google Scholar] [CrossRef]
  244. Xin, S.H.; Tan, L.; Cao, X.; Yu, J.T.; Tan, L. Clearance of Amyloid Beta and Tau in Alzheimer’s Disease: From Mechanisms to Therapy. Neurotox. Res. 2018, 34, 733–748. [Google Scholar] [CrossRef]
  245. Tsigelny, I.F.; Crews, L.; Desplats, P.; Shaked, G.M.; Sharikov, Y.; Mizuno, H.; Spencer, B.; Rockenstein, E.; Trejo, M.; Platoshyn, O.; et al. Mechanisms of hybrid oligomer formation in the pathogenesis of combined Alzheimer’s and Parkinson’s diseases. PLoS ONE 2008, 3, e3135. [Google Scholar] [CrossRef]
  246. Twohig, D.; Nielsen, H.M. α-synuclein in the pathophysiology of Alzheimer’s disease. Mol. Neurodegener. 2019, 14, 23. [Google Scholar] [CrossRef]
  247. Danysz, W.; Parsons, C.G.; Möbius, H.J.; Stöffler, A.; Quack, G. Neuroprotective and symptomatological action of memantine relevant for Alzheimer’s disease—a unified glutamatergic hypothesis on the mechanism of action. Neurotox. Res. 2000, 2, 85–97. [Google Scholar] [CrossRef]
  248. Mintun, M.A.; Lo, A.C.; Duggan Evans, C.; Wessels, A.M.; Ardayfio, P.A.; Andersen, S.W.; Shcherbinin, S.; Sparks, J.; Sims, J.R.; Brys, M.; et al. Donanemab in Early Alzheimer’s Disease. N. Engl. J. Med. 2021, 384, 1691–1704. [Google Scholar] [CrossRef]
  249. Fields, C.R.; Bengoa-Vergniory, N.; Wade-Martins, R. Targeting Alpha-Synuclein as a Therapy for Parkinson’s Disease. Front. Mol. Neurosci. 2019, 12, 299. [Google Scholar] [CrossRef]
  250. Maiti, P.; Manna, J.; Dunbar, G.L. Current understanding of the molecular mechanisms in Parkinson’s disease: Targets for potential treatments. Transl. Neurodegener. 2017, 6, 28. [Google Scholar] [CrossRef]
  251. Politis, M.; Niccolini, F. Serotonin in Parkinson’s disease. Behav. Brain Res. 2015, 277, 136–145. [Google Scholar] [CrossRef]
  252. Grosch, J.; Winkler, J.; Kohl, Z. Early Degeneration of Both Dopaminergic and Serotonergic Axons—A Common Mechanism in Parkinson’s Disease. Front. Cell Neurosci. 2016, 10, 293. [Google Scholar] [CrossRef] [PubMed]
  253. Mallajosyula, J.K.; Kaur, D.; Chinta, S.J.; Rajagopalan, S.; Rane, A.; Nicholls, D.G.; Di Monte, D.A.; Macarthur, H.; Andersen, J.K. MAO-B elevation in mouse brain astrocytes results in Parkinson’s pathology. PLoS ONE 2008, 3, e1616. [Google Scholar] [CrossRef] [PubMed]
  254. Datta, P.; Rewers-Felkins, K.; Baker, T.; Hale, T.W. Transfer of Montelukast into Human Milk During Lactation. Breastfeed. Med. 2017, 12, 54–57. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Leukotriene biosynthesis pathway and receptor recognition. Leukotrienes are synthesised upon activation of the immune system through an LT biosynthesis cascade, acting on various organs through different receptors. V is a 5-LOX inhibitor, whereas W, X, Y, and Z are inhibitors of the leukotriene pathways; all are marked in red. Image adapted from Funk [19]. Created with BioRender.com.
Figure 1. Leukotriene biosynthesis pathway and receptor recognition. Leukotrienes are synthesised upon activation of the immune system through an LT biosynthesis cascade, acting on various organs through different receptors. V is a 5-LOX inhibitor, whereas W, X, Y, and Z are inhibitors of the leukotriene pathways; all are marked in red. Image adapted from Funk [19]. Created with BioRender.com.
Pharmaceuticals 15 01039 g001
Figure 3. Positive and negative aspects of neuroinflammation. The consequences of neuroinflammation depend on the duration and severity of the immune response. (Left) induction of sickness behaviour to restore the host’s homeostasis after infection; (right) chronic neuroinflammation tends to carry negative consequences. Low, med, and high refer to a low, medium, and high levels of inflammation. Image adapted from DiSabato et al. [68], with permission from John Wiley and Sons. Created with BioRender.com.
Figure 3. Positive and negative aspects of neuroinflammation. The consequences of neuroinflammation depend on the duration and severity of the immune response. (Left) induction of sickness behaviour to restore the host’s homeostasis after infection; (right) chronic neuroinflammation tends to carry negative consequences. Low, med, and high refer to a low, medium, and high levels of inflammation. Image adapted from DiSabato et al. [68], with permission from John Wiley and Sons. Created with BioRender.com.
Pharmaceuticals 15 01039 g003
Figure 4. Leukotrienes in the central nervous system: a pleiotropic effect. Broken lines symbolize putative actions of leukotriene signalling that require further investigations. Image adapted from Michael et al. [67], with permission from Elsevier. Created with BioRender.com.
Figure 4. Leukotrienes in the central nervous system: a pleiotropic effect. Broken lines symbolize putative actions of leukotriene signalling that require further investigations. Image adapted from Michael et al. [67], with permission from Elsevier. Created with BioRender.com.
Pharmaceuticals 15 01039 g004
Table 1. Contribution of different CYP isoforms to MTK metabolism in human liver microsomes. The colour gradient indicates the relative contribution of each CYP isoform: the darker the shade, the more relevant is the CYP isoform role.
Table 1. Contribution of different CYP isoforms to MTK metabolism in human liver microsomes. The colour gradient indicates the relative contribution of each CYP isoform: the darker the shade, the more relevant is the CYP isoform role.
Recombinant CYP Isoform
1A22A62B62C82C92C192D62E13A43A5
M2a [121]
[123]
[124] *
[122] *
M2b [121]
[123]
M3 [121]
[123]
[124]
M4 [123]
M5a [121]
[123]
[124]
[122] δ
M5b [121]
[123]
[124]
M6 [121]
[123]
[124]
[122]
Metabolite formation rate (no shade—lower; darker shade—higher)
Not included in the study
* No distinction between M2a and M2b diastereomers was made. δ No difference between M5a and M5b was reported.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Marques, C.F.; Marques, M.M.; Justino, G.C. Leukotrienes vs. Montelukast—Activity, Metabolism, and Toxicity Hints for Repurposing. Pharmaceuticals 2022, 15, 1039. https://doi.org/10.3390/ph15091039

AMA Style

Marques CF, Marques MM, Justino GC. Leukotrienes vs. Montelukast—Activity, Metabolism, and Toxicity Hints for Repurposing. Pharmaceuticals. 2022; 15(9):1039. https://doi.org/10.3390/ph15091039

Chicago/Turabian Style

Marques, Cátia F., Maria Matilde Marques, and Gonçalo C. Justino. 2022. "Leukotrienes vs. Montelukast—Activity, Metabolism, and Toxicity Hints for Repurposing" Pharmaceuticals 15, no. 9: 1039. https://doi.org/10.3390/ph15091039

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop