Skip to main content
Current Neuropharmacology logoLink to Current Neuropharmacology
. 2021 May 27;19(6):885–895. doi: 10.2174/1570159X18666200924122732

Inflammation Drives Alzheimer's Disease: Emphasis on 5-lipoxygenase Pathways

Aisha Siddiqui 1, Sayeed Akhtar 3,*, Zahoor Shah 2, Iekhsan Othman 1, Yatinesh Kumari 1,*
PMCID: PMC8686299  PMID: 32972344

Abstract

It is a known fact that inflammation affects several physiological processes, including the functioning of the central nervous system. Additionally, impairment of lipid mechanisms/pathways have been associated with a number of neurodegenerative disorders and Alzheimer’s Disease (AD) is one of them. However, much attention has been given to the link between tau and beta-amyloid hypothesis in AD pathogenesis/prognosis. Increasing evidences suggest that biologically active lipid molecules could influence the pathophysiology of AD via a different mechanism of inflammation. This review intends to highlight the role of inflammatory responses in the context of AD with the emphasis on biochemical pathways of lipid metabolism enzyme, 5-lipoxygenase (5-LO).

Keywords: Alzheimer's Diseases, 5-lipoxygenase, Neuro Inflammation, Neurodegeneration, Lipid Molecule, Arachidonic Acid

1. INTRODUCTION

Alzheimer’s Disease (AD) is a destructive neurodegenerative disease and a leading cause of dementia in the elderly population that declines memory functions, decision making, reasoning ability, weakens cognition, and causes changes in behavior. In 2019, it was estimated by Alzheimer’s disease international that 50 million people are living with dementia, a figure which may rise up to 152 million by the year 2050 [1].

The two classic major pathological hallmarks of AD include amyloid-beta (Aβ) accumulation and neurofibrillary tangles (NFTs) formation [2]. Some new pathogenic biomarkers have shown up which makes it even more challenging to slow down the progression of AD. Soluble amyloid β peptide (Aβ) is build up at an early stage that initiates various faulty cycles like a decline in synaptic communication, ruptures mitochondria, and enhances oxidative stress that causes sustained endoplasmic reticulum (ER) related stress reaction [3]. Other pathological hallmarks of AD, which were initially described by Alois Alzheimer, is the higher occurrence of ‘lipoid granules’ or ‘adipose inclusions’ suggesting dysregulated lipid metabolism [4]. Inflammation, on the other hand, is a component of initial innate response that en-gages immune cells to the spot of stress mainly through the chemotactic function of activated chemokines. Furthermore, the dual involvement of chemokines as pro and anti-inflammatory is well evident and must be considered for both clinical and immunopathological approaches in AD [5, 6]. Eicosanoids are the signaling molecules and inflammatory mediators that are generated in the pathway of Arachidonic Acid (AA) metabolism mediated by 5 lipoxygenases (5-LOs) [7]. These biologically active lipid molecules could manipulate the working of the central nervous system (CNS) as well as the pathophysiology of neurodegenerative disorders, such as AD, via various mechanisms involved in classical inflammation and are discussed here.

2. ALZHEIMER’S DISEASE AND AGING

A large number of cases of AD are sporadic, without any regular genetic expression among different cases. In 2019, Alzheimer’s Association stated that the signs and symptoms of AD typically become diagnosable only between the age of 65-74 years (3%), 75-84 years (17%), and ≥ 85 years (32%) [1, 2].

Generally, the aging brain is correlated with the high risk of chronic neurodegenerative mechanisms involved in disease progression [8, 9]. In this view, the biochemical pathways suggest that brain aging is associated with the activation of microglia and persistent brain inflammation results in neuronal death [10]. Patients with AD go through remarkable disintegration of neurons, as a result of an extracellular aggregation of Aβ peptide and intracellular aggregation of tau protein that leads to the formation of neurofibrillary tangles [11, 12]. Additionally, it has become evident by the different published studies that levels of 5-lipoxygenase(5-LO) and its metabolites increase age dependently [13, 14]. Therefore, aging and involvement of 5-LO have been considered as potential risk elements for the progression of AD [15, 16].

3. MECHANISM INVOLVED IN THE DEVELOPMENT OF ALZHEIMER’S DISEASE

Scientific developments over the past decades have expanded the understanding of cellular and molecular aspects of AD (Fig. 1). In spite of that, AD remains mainly idiopathic, therefore, treatments and therapies meant to battle the disease progression successfully are still lacking. Studies understanding the initiation, progression, and prevention of AD and its related conditions are limited. Emerging data indicates that the healthy physiological interactions between neurons, astrocytes, microglia, as well as vascular cells are prerequisites for proper brain functioning. Early diagnosis and therapy can help in protecting neural circuits from damage, but it may get worse if not treated properly [17].

Fig. (1).

Fig. (1)

Association of 5-Lipoxygenase enzyme (5-LO) in progression of Alzheimer’s disease. FLAP- 5-Lipoxygenase activating protein, IL- Interleukin, TNFα- Tumour Necrosis Factor alpha, APP- Amyloid Precursor Protein. (A higher resolution / colour version of this figure is available in the electronic copy of the article).

3.1. Amyloid β Plaques

The irregular breakup of Aβ protein is involved in Aβ related pathology that gives rise to monomeric form and later on accumulates to form oligomeric Aβ. These Aβ monomers combine with each other and form Aβ plaques and fibrils [18]. The role of Amyloid precursor protein (APP) is still unclear, but it is expected to be responsible for cell growth [19]. Generally, non-amyloidogenic APP proteolysis takes place with the help of α- secretase or γ-secretase resulting in the formation of dissoluble fragments [20]. Whenever γ secretase and faulty β-secretase over-activate the cleavage of APP, it results in the formation of insoluble peptides of Aβ, which accumulate to form Aβ plaques in the brain and outside the neurons [21-26]. The exact function of Aβ in the pathology of AD is still an unsettled question as plaques of Aβ may aggregate for up to 10 years without any detectable symptoms or diagnosis of AD, thus, being the reason for the development of AD in the aged population.

3.2. Tau Protein and Neurofibrillary Tangles

The presence of NFTs emerges mainly after the tau hyperphosphorylation. It is a microtubule linked protein that secures microtubule and its hyperphosphorylation initiated AD pathology [27, 28]. In the case of AD, phosphorylation of tau protein takes place at numerous sections leading to the elimination of tau from the microtubule. This results in the shrinking of microtubule and interrupting various cellular functions [29-31]. Moreover, hyper-phosphorylation of tau results in its accumulation into coiled fragments that ultimately give rise to NFTs [32-36]. Tau tangles aggregation and the weakened cellular actions cause failure in neuronal structure and nutrient transport, leading to the death of neurons [30]. So far, major approaches were made to target the inhibition of Aβ plaques and NFTs, which failed in clinical trials. Therefore, effective treatment modalities targeting the pathology of AD remain to be resolved and revisited. Thus, regardless of the broad and dynamic studies, inspecting the mechanisms culpable for different pathologies is required due to the significant gap in awareness of AD pathology [37]. The first pathology, as discussed previously based on the Aβ plaques, may accumulate for up to or more than 10 years without any signs/symptoms of AD before its precise diagnosis [38, 39]. NFTs hypothesis reviewed already explains, the total load of the tangles is associated with the decline of cognition in AD. Anyhow, the presence of NFTs is detected before the initiation of AD symptoms in both preclinical studies and non-symptomatic AD cases [40-42].

3.3. Role of Inflammatory Responses

In the past ten years, a third basic pathological element of AD has appeared that may be fruitful in understanding the pathogenesis of AD and is also associated with the above two basic pathologies. Studies have demonstrated that in AD patient’s brain, not only Aβ plaques and NFTs are found, but there is also a constant inflammation led presence of inflammatory cytokines, such as Interleukin-6 (IL-6), Interleukin-1β (IL-1β), and Tumour necrosis factor-α (TNF-α) [43-45]. Various studies have emerged to investigate the inflammatory response in post-mortem tissues of AD patients and are now regularly looked at in AD-related preclinical studies [46, 47].

Anti-inflammatory cytokines (IL-4, IL-10, and IL-13) can activate M2 subtype microglia cells, surrounding amyloid neuritic plaques and can also regulate Aβ-induced production of the inflammatory cytokines. Recent data states that the level of pro-inflammatory cytokines remains increased in AD brains. IL-1, IL-10, and IL-13, along with TNF-α is a potent inflammatory cytokine that promotes the pathophysiological process and triggers signaling pathways that can regulate various cellular mechanism linked with gene expression, cell viability, synaptic integrity, and ion homeostasis [6].

Defense mechanism against toxins, infection, and injury is well understood in case of acute inflammation in the brain. However, any imbalance between pro-inflammatory and anti-inflammatory responses leads to chronic inflammation, which then further activates microglial cells and releases cytokines [48-51]. Microglia are the inhabitants of immune cells inside the CNS [52]. Microglial cells remain dormant, a ‘relaxing’ state in healthy brains, and are identified as branched cells with small somas [53, 54]. In this form, these cell somas remain static. However, these soma cells prepare themselves to expand and retract, inspecting their surroundings and linking with astrocytes and neurons [55-57]. They serve as the first line of defense systems whenever there is a threat of pathogens or injury to the brain. Microglia get activated in neurodegenerative diseases, tumors, and stroke, and these cells encircle dead or damaged cells and make area free from cellular debris, a phenotype known as phagocytic macrophages [58]. In AD, it is predicted that the existence of Aβ results in the activation of microglial cells and these microglia then move towards plaques and phagocytose the Aβ [59-61]. The constant activation/reactivation leads to a decline in the microglial binding and Aβ phagocytosing activity. In the milieu, the degradation of microglia leading enzyme activity conclusively reduces the breakdown of Aβ plaques [62, 63]. Apart from the microglia, astrocytes are also considered as one of the most important members for the degradation and clearance of Aβ. This serves as a trophic aid to neurons and maintains the protective boundary between neurons and amyloid deposits [64, 65]. In AD, the existence of large numbers of astrocytes correlated with Aβ deposits if these would produce certain molecules that help in the recruitment of astrocytes. However, it is evidenced that engaged astrocytes gather at Aβ plaque vicinity, leading to neuroinflammation and subsequent neurotoxicity induced by NO in the presence of L-arginine delivering enzyme argininosuccinate synthetase and inducible nitric oxide synthase (iNOS) [66]. Another report proposed that astrocytes may be responsible as an origin of Aβ because they reveal an excessive amount of β-site APP cleaving enzyme 1 (BACE1) as feedback to chronic stress [65].

4. 5-LIPOXYGENASE AND INFLAMMATION

5-LO is a monomer protein comprising of 673 amino acids and has been identified in various distinct mammalian species. Lipoxygenases (LO) are a cluster of iron-enclosed dioxygenases that catalyse oxidation reaction and add oxygen to AA, docosahexaenoic acid (DHA), and various polyunsaturated fatty acids (PUFA). LO enzymes are found in different isoforms depending upon the tissue type and their location, like epidermis type (LOX-3) or reticulocyte type (LOX-15). Some LO enzymes catalyze few reactions, such as the addition of molecular oxygen to AA at carbon 5, 12 and 15 in different ratios in reticulocytes. This results in the production of 5-, 12-, 15- hydroperoxy- eicosatetraenoic acid (HPETE) as an end product [67, 68]. Overall, there are five isoforms of LOs that are expressed in humans/animals: 15-LO, 12-LO, 3-LO, 8-LO, and 5-LO. This naming of the LOs is based on the position of oxygen added into substrate like AA [69]. These lipid mediators play an important role in the oxidative degradation of lipids, converting it into the peroxide as they exist in animals as well as in vegetal kingdoms [70, 71]. Among all the LOX enzymes, 5-LO is crucial which helps in the conversion of AA to 5(S)-HPETE and leukotrienes (LTs). Inflammatory mediators like LTs and inflammatory eicosanoids act on different receptors and persuade various responses like leukocytes chemotaxis and enhanced vascular permeability as well as several effects in neurons that are even not well known [72-74]. The LTs that remain present in the brain play a significant role in pathological alterations in brain tissues [75, 76]. However, lag time is required for the LO enzymes to activate the inactive ferrous form of the enzyme to the active ferric form. Even though the analytical performance of the LOs is still argumentative, its radical character is generally determined. Three stages are involved in sequence: (1) abstraction of stereo-selective hydrogen from a two allelic methylene group, (2) rearrangement of radicals, and (3) stereo-specific infusion of the oxygen molecule and decline of hydroperoxy-radical intermediate to the analogous anion [68].

5. 5-LIPOXYGENASE AND CATALYTIC PATHWAYS

Figure 2 illustrates the role of 5-LO and its related catalytic pathways involved in the inflammation. AA is one of the main free fatty acids (FFA) produced by the metabolism of cytosolic phospholipase A2 (cPLA2). In the initial two steps of biosynthesis, AA is oxidized at Carbon-5 to produce 5-HPETE in the presence of FLAP and 5-LO pursued by successive dehydration of hydroperoxide, yielding to an intermediate product LeukotrieneA4 (LTA4) and 5-HETE. Relying on the accessible enzymes, the extremely unsteady epoxide LTA4 either hydrolyzed to LeukotrieneB4 (LTB4) in the presence of LTA4 hydrolase or get associated to glutathione to produce LeukotrieneC4 (LTC4) with the help of LTC4 synthase. The compound LTC4 is further metabolized to LeukotrieneD4 (LTD4) and LeukotrieneE4 (LTE4) after successful termination of glycine and glutamic acid by glutamyl transferase and LTD4 dipeptidase [77, 78]. LTB4 receptors include Leukotriene B4 receptor 1 (BLT1) and Leukotriene B4 receptor 2 (BLT2), Cysteinyl leukotriene receptor 1 (CysLT1), and Cysteinyl leukotriene receptor 2 (CysLT2), which are G-protein coupled receptors and contribute to inflammation [79].

Fig. (2).

Fig. (2)

Role of 5-Lipoxygenase enzyme (5-LO) in inflammation- Arachidonic acid is the primary substrate which is produced by metabolism of cytosolic phospholipase A2 (cPLA2). 5-LO acts on arachidonic acid to releases 5 HPETE which metabolizes to form Leukotrienes (LTA4) and 5-HETE. Furthermore, this leukotriene act on hydrolase and synthase to produce other leukotrienes (LTB4, LTC4). LTB4 bind to their respective receptors which are G-Protein coupled receptors and contributes in inflammation. Leukotrienes LTC4 by incorporating γ Glutamyl transferase-1 pursue to form LTD4 which act on LTD4 dipeptidase to generate LTE4. These leukotrienes collectively called as cysteinyl leukotrienes they bind to their receptors (CysLT1, CysLT2) which are again GPCRs and assist in Chemokine production, Immune cell activation and Inflammation. Whereas, 5-HETE which is produced from 5-HPETE in the presence of 5-LO binds to eicosanoid receptors to trigger inflammation. (A higher resolution / colour version of this figure is available in the electronic copy of the article).

On the other hand, inflammatory mediators that are produced by the 5-LO act on the Oxoeicosanoid receptors. This includes eicosanoids 5-HETE and 5-Oxo-eicosatetraenoic acid (5-oxo-ETE). The metabolites of 5LO activation produce LTs and 5HETE that trigger chemotaxis of immune cell and are known to be a key player in the pathophysiology of inflammation in different diseases like asthma and allergy [80]. Importantly, the expression of 5LO and its metabolic products increase with an increase in the age of both animals and humans. The most vulnerable areas like the cortex and hippocampus are specifically sensitive to neurodegeneration and are usually found to be rich in 5LO protein levels in AD [81, 82]. There are numerous elements required by 5LO to become effective and concurrent to upsurge cellular calcium ions that motivate the transfer of the enzyme to the membrane from the cytosol. 5LO, in the membrane, is combined with the integral membrane protein, which is commonly known as FLAP [83, 84]. Apart from oxidation, 5-LO and its activating protein FLAP play a major role in initiating inflammation in AD. Interruption in these pathways lowers the microglia as well as astrocytes in the animals with AD, as evident by the immunohistochemical analyses [85]. In addition, there is a correlated reduction in the levels of pro-inflammatory cytokines with the disruption in 5-LO or FLAP. In the brain ischemia-reperfusion model, it was reported that the translocation of neuronal 5-LO from the cytosol to the membrane was followed by the enhanced formation of LTs [86]. Therefore, inhibition of 5-LO reduces neurodegeneration in the ischemic brain [87].

6. 5-LIPOXYGENASE AND ALZHEIMER’S DISEASE

Various studies strengthen the involvement of 5-LO enzymes in the development of AD. This involves a comparison between the post-mortem studies of healthy control and AD brains and demonstrated that the levels of 5-LO were higher in the hippocampus and cortex regions of AD patient’s brain. However, no significant change was observed when the assay was carried out between the cerebellum (a region lacking AD pathology) of both the brains of healthy control and AD [88, 89]. A genetic study conducted on humans revealed that 5-LO gene polymorphism is linked to early and late development of AD. However, large-scale populace studies are yet to affirm these findings. A separate study examined the epigenetic flaw of 5-LO in mononuclear peripheral blood cells of individuals with late development of AD and age-balanced controls. This demonstrated a remarkable increase in the expression of the 5-LO gene in AD patients as compared to the control ones [90-93].

The small protein FLAP triggers the 5-LO enzyme, leading to a complex formation, which conveniently produces 5- HPETE from AA that gets converted to LTs. FLAP remains on the nuclear membrane to behave as a conveyor of LOs [94]. It seems that both 5-LO and FLAP show a reduction in the fundamental pathology of AD. At the same time, they reduce the oxidative stress and inflammation induced by AD pathology. The inhibition of the 5-LO pathway in cultured hippocampal neurons of the rat demonstrated the decrease in reactive oxygen species (ROS) formation, induced by antibodies and successive calcium dysregulation in the concentration-controlled process [95]. Interruption in the 5-LO pathway prevented from glutamate-induced excitation toxicity in rats, particularly in older animals [96]. in vitro studies evidenced that over-expression of 5-LO does not result in oxidative stress itself, but when over-expressed in the presence of Aβ peptides, it lowers the levels of glutathione peroxidase and catalase enzymes [97]. This was observed that the inhibition in the pathway of 5-LO by a target inhibitor, pyrazole CNB-00, prevents the endoplasmic reticulum (ER) abnormality and toxicity of proteasome, promoted by Aβ peptide present in cultured neurons and in-vivo study [98]. Surprisingly, in in-vitro studies, blockage in FLAP is insufficient to shield against oxidative toxicity induced by the peptide Aβ. Evidences suggest that even after the decoupling of 5-LO from Aβ peptide metabolism, its capability to introduce molecular oxygen remains protected and preventing pro-oxidative characteristics in a LTs independent manner. However, the fact is not well understood, and therefore, more research must be carried out to investigate this phenomenon further. Moreover, inhibition of LTs is associated with preventing cognitive loss provoked by traumatic injury of the brain in rats due to the genesis of oxidative stress that is a well-established risk element for AD [99]. FLAP protein targeted drugs have already been used in humans. The agent, DG-031reduces the risk of myocardial infarction by lowering the release of LTB4 [100]. An arthritis model showed that the mice lacking arachidonate 5-lipoxygenase activating protein (ALOX5ap), a protein-coding gene, does not initiate antibody accumulation against collagen. Thus, it represents that 5-LO controls inflammation without disturbing immune response [101]. Mice lacking ALOXap shows recovered AD like symptoms [102, 103]. Above evidences indicated that FLAP and 5-LO are together involved in the LTs pathway in neurological disorders.

LO is not only responsible for the synthesis of active lipids but also plays an essential role in oxidizing the lipid segments in the cell membrane. It produces the activators, causing structural changes and leading to maturation and differentiation of a variety of cells [104]. The mouse has a total of seven different Los and among these, five different LO has also been found in humans. This includes 5-LO, 12-LO (platelet-type and leucocyte-type), and 15-LO, which is divided into subtypes 15-LO-1 (leucocyte-type) and 15-LO-2 (epidermis type) [105, 106].

There are various tissues in which LOs are expressed and involved in different diseases like diabetes (both type1 and type 2), kidney diseases, atherosclerosis, and obesity [107, 108]. Currently, LOs are expected to play a key role in the development of neuronal diseases, including AD [90].

Arachidonate 5-Lipoxygenase (ALOX5) has also shown to play a prominent role in mediating inflammation in the brain. Cannabinoid Receptor 2 activation down-regulates 5-lipoxygenase (Alox5) expression by suppressing the JNK/c-Jun activation [109, 110].

Besides asthma and inflammatory reactions, the role of 5-LO in the induction or progression of AD is well evident. In AD, ALOX5 is overexpressed and its importance in neurodegenerative diseases needs to be discussed at least in brief [111]. Current research states that ALOX5 lacking mice show defensive action against anxiety, which may be due to the modification in neuronal function by ALOX5 [112]. Consecutive research was performed on a transgenic mouse model of AD, which described the efficacy of 5-LO inhibitor (Zileuton) and predicted that ALOX5 is either involved in the initiation or progression of AD [113].

To understand the role of LOs profoundly, a study in 2004 expressed the very first data that exposed the modified role of 12-LO in AD using post-mortem autopsy brain analysis [114]. The study highlights that levels of 12/15-LO protein are increased in the temporal and frontal cortex. This rise is a specific hallmark of oxidative degradation of lipids, isoprostane 8, 12-iso-iPF2aVI, is and conversely associated with the amount of vitamin E, indicating a notable aspect of 12/15-LO acting as an oxidative stressor. Subsequently, the excitement of 12/15-LO leads to the excess production of 12/15-HETE in these cortexes, which are parallel with lipid peroxidation along with the observation of microtubule-associated protein tau (MAPT) protein in the cerebrospinal fluid of patients with AD [115].

Furthermore, the elevated levels of 12/15-HETE were also observed in the cerebrospinal fluid of the people with mild cognitive defects, stating that 12/15-LO performs a critical role during the progression and in the initial phase of AD. In an agreement, the lack of 12/15-LO reduces oxidative stress in the brains of mice deficient with apolipoprotein E (ApoE) [116].

7. CURRENT STATUS OF 5 LIPOXYGENASE IN ALZHEIMER’S DISEASE

The epidemiological study indicated that patients with AD would be a future burden and an enormous national health threat. In approaching the national health challenge of AD, currently, narrow options of AD therapy are available. However, few medications were made available to the public after phase II and phase III trials that either inhibit the enzyme acetylcholinesterase or target N-methyl D-aspartate (NMDA). But, most of these drugs are used for symptomatic treatment and are not a promising regimen in the treatment of AD as their fate is not clear [4, 7]. AD is considered a burden worldwide and there is a shortage of adequate therapeutic agents, therefore, target-oriented research that effectively acts on AD pathophysiology needs to be prioritized to settle this malady. Many studies have been performed to reveal the role of LO inhibitors in AD [117, 118]. Selective inhibition is found to be challenging because of the existence of many isoforms, and this narrows the successful development of therapeutic agents. Clinically approved drug-like zileuton is an anti-asthmatic drug which inhibits 5-LO. The research has been conducted targeting FLAP for making changes in 5-LO in pathophysiological surroundings. Fiboflapon is another clinically approved FLAP inhibitor that limits LTB4 production and other LTs [119]. The curative effects of another drug (NCT01147744) have been confirmed in adults and juveniles with stubborn asthma [120]. Other FLAP inhibitors like quiflapon (MK- 591), veliflapon (DG-031), AZD6642, BRP-7, and BRP-187 are in clinical trials. The only FLAP inhibitor, which successfully finished phase II clinical trials in asthma, to date, is GSK2190918 [121]. Apart from targeting the 5-LO and FLAP, there is another study that focuses on NACHT (NTPase domain), LRR (Leucine-rich repeat), and PYD (Pyrin) domains containing protein 3, collectively called as cryopyrin. They remain encoded by the gene NLRP3 (nucleotide-binding domain like receptor protein 3). Till now, the discussion has been made on the role played by Aβ activation that is directly responsible for the progression of AD. But different studies state that Aβ ameliorates AD indirectly by activating NLRP3 inflammasome, which carries NLRP3, caspase-1, and ASC (Apoptosis-associated speck like protein containing a caspase). In the Aβ plaques of AD models of mice and also in AD patients, levels of the cleaved and activated caspase-1 were found to be elevated [122, 123]. As intracellular deposition of tau is not a ruling signature of diagnosis, tauopathy is also considered to be secondary in terms of AD. Fundamental tauopathy, like frontotemporal dementia (FTD), also shows cognitive defects along with neuroinflammation [124].

8. FUTURE POSSIBILITIES IN ALZHEIMER’S DISEASE

AD is one of the age-related progressive brain illnesses among the elderly population. The aforementioned report enlightened the role of inflammation played in the progression of AD, apart from Aβ and tau involvement and also highlighted the other elements that contribute to AD pathology. It is evident through literature that LO plays an important role in AD pathogenesis. Therefore, target-oriented investigation is the need of the hour. Although there are marketed preparations available for the inhibition of LO enzymes, natural compounds need more attention for therapy development. The diagnosis of AD is also insufficient and needs to be addressed to develop biomarker kits that will detect the early onset of AD. Usually, at the time of diagnosis of AD, the disease progression to advanced stage is always a concern. There is a need to investigate prognostic biomarkers of inflammation that will help the researchers in predicting the disease before it starts which is a greater challenge. Additionally, large scale investigation is required in AD patients and preclinical animal models to further explore the microglial signaling pathway and NLRP3 inflammasome activation.

CONCLUSION

In the last decade, understanding of various pathological changes in AD has been greatly expanded. However, target-based treatment is still lacking and needs to be discussed thoroughly. The pathological alterations in the brain due to AD include neurodegeneration, extracellular accumulation of Aβ plaques, and formation of intracellular NFTs of hyperphosphorylated protein Tau. The alteration of homeostasis between inflammatory (IL-1, IL-10, IL-13 and TNF-α) and anti-inflammatory cytokines (IL-4, IL-10, and IL-13) promotes the pathophysiological damages and further triggers different signaling pathways that can regulate other cellular mechanisms linked with gene expression, cell viability, synaptic integrity, and ion homeostasis. Along with this, microglia (a glial cell type), that remain present in the brain and spinal cord, act as primary resident immune cell. Inflammatory responses due to neurodegeneration further activate these microglia and clear the debris, including Aβ, by phagocytosis. Also, astrocytes gather at the surroundings of Aβ and further produce neuroinflammation and related toxicity.

Importantly, with these pathological changes, 5-LO mediated neuroinflammation is a major area of concern in the development of AD. FLAP is the small protein that triggers the 5-LO enzyme that produces HPETE and LTs from AA. Recent evidence undoubtedly suggests that the involvement of 5-LO promotes both the initiation and progression of disease in elderly age. In addition, different eicosanoids like LTs are generated by AA metabolism mediated by 5-LO and act as inflammatory mediators. This initiates chemotaxis and enhances vascular permeability in the neuronal cells. The increased expression of 5-LO and its related metabolic products with age are well evident. In a similar context, the interruption of 5-LO mediated pathways reduces the number of microglia as well as astrocytes, indicating reduced neuroinflammation.

Based on these opportunities, many studies have been conducted to develop 5-LO inhibitors in AD. However, selective inhibition is an area of great concern due to the presence of many isoforms of lipoxygenase. Undoubtedly, we need to explore all the different corners of 5-LO targeted investigation to add a milestone in the treatment of AD.

ACKNOWLEDGEMENTS

Declared none.

CONSENT FOR PUBLICATION

Not applicable.

FUNDING

We thank Tropical Medicine and Biology (TMB) Multidisciplinary Platform (TMB-2021-SG3185140921-YatiKu), Monash University Malaysia, and Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, for the financial support to our work on the role of dietary lipoxygenase antagonist in AD, which was the starting point of this review article.

CONFLICT OF INTEREST

The authors have no conflicts of interest, financial or otherwise.

REFERENCES

  • 1.International, A. s. D., World Alzheimer report 2019: attitudes to dementia. UK: Alzheimer’s Disease International London; 2019. [Google Scholar]
  • 2.Gaugler J., James B., Johnson T., Marin A., Weuve J. 2019 Alzheimer’s disease facts and figures. Alzheimers Dement. 2019;15(3):321–387. [Google Scholar]
  • 3.Murphy K.E., Park J.J. Can co-activation of Nrf2 and neurotrophic signaling pathway slow Alzheimer’s disease? Int. J. Mol. Sci. 2017;18(6):1168. doi: 10.3390/ijms18061168. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Wong M.W., Braidy N., Poljak A., Pickford R., Thambisetty M., Sachdev P.S. Dysregulation of lipids in Alzheimer’s disease and their role as potential biomarkers. Alzheimers Dement. 2017;13(7):810–827. doi: 10.1016/j.jalz.2017.01.008. [DOI] [PubMed] [Google Scholar]
  • 5.Azizi G., Khannazer N., Mirshafiey A. The potential role of chemokines in Alzheimer’s disease pathogenesis. Am. J. Alzheimers Dis. Other Demen. 2014;29(5):415–425. doi: 10.1177/1533317513518651. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Azizi G., Mirshafiey A. The potential role of proinflammatory and antiinflammatory cytokines in Alzheimer disease pathogenesis. Immunopharmacol. Immunotoxicol. 2012;34(6):881–895. doi: 10.3109/08923973.2012.705292. [DOI] [PubMed] [Google Scholar]
  • 7.Tan C-C., Yu J-T., Wang H-F., Tan M-S., Meng X-F., Wang C., Jiang T., Zhu X-C., Tan L. Efficacy and safety of donepezil, galantamine, rivastigmine, and memantine for the treatment of Alzheimer’s disease: a systematic review and meta-analysis. J. Alzheimers Dis. 2014;41(2):615–631. doi: 10.3233/JAD-132690. [DOI] [PubMed] [Google Scholar]
  • 8.McGeer P.L., McGeer E.G. Inflammation and the degenerative diseases of aging. Ann. N. Y. Acad. Sci. 2004;1035(1):104–116. doi: 10.1196/annals.1332.007. [DOI] [PubMed] [Google Scholar]
  • 9.Zhao Y., Zhao B. Oxidative stress and the pathogenesis of Alzheimer's disease. Oxid. Med. Cell. Long. 2013;2013:316523. doi: 10.1155/2013/316523. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Kirkwood T.B., Feder M., Finch C.E., Franceschi C., Globerson A., Klingenberg C.P., LaMarco K., Omholt S., Westendorp R.G. What accounts for the wide variation in life span of genetically identical organisms reared in a constant environment? Mech. Ageing Dev. 2005;126(3):439–443. doi: 10.1016/j.mad.2004.09.008. [DOI] [PubMed] [Google Scholar]
  • 11.Hou Y., Dan X., Babbar M., Wei Y., Hasselbalch S.G., Croteau D.L., Bohr V.A. Ageing as a risk factor for neurodegenerative disease. Nat. Rev. Neurol. 2019;15(10):565–581. doi: 10.1038/s41582-019-0244-7. [DOI] [PubMed] [Google Scholar]
  • 12.Wyss-Coray T. Ageing, neurodegeneration and brain rejuvenation. Nature. 2016;539(7628):180–186. doi: 10.1038/nature20411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Bachiller S., Jiménez-Ferrer I., Paulus A., Yang Y., Swanberg M., Deierborg T., Boza-Serrano A. Microglia in neurological diseases: a road map to brain-disease dependent-inflammatory response. Front. Cell. Neurosci. 2018;12:488. doi: 10.3389/fncel.2018.00488. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Di Francesco A., Arosio B., Gussago C., Dainese E., Mari D., D’Addario C., Maccarrone M. Involvement of 5-lipoxygenase in Alzheimer’s disease: a role for DNA methylation. J. Alzheimers Dis. 2013;37(1):3–8. doi: 10.3233/JAD-130506. [DOI] [PubMed] [Google Scholar]
  • 15.Chu J., Pratico D. The 5-lipoxygenase as a common pathway for pathological brain and vascular aging. Cardiovasc. Psychiat. Neurol. 2009;2009:174657. doi: 10.1155/2009/174657. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Praticò D. Oxidative stress hypothesis in Alzheimer’s disease: a reappraisal. Trends Pharmacol. Sci. 2008;29(12):609–615. doi: 10.1016/j.tips.2008.09.001. [DOI] [PubMed] [Google Scholar]
  • 17.Fakhoury M. Microglia and astrocytes in Alzheimer’s disease: implications for therapy. Curr. Neuropharmacol. 2018;16(5):508–518. doi: 10.2174/1570159X15666170720095240. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Manev H., Manev R. 5-Lipoxygenase (ALOX5) and FLAP (ALOX5AP) gene polymorphisms as factors in vascular pathology and Alzheimer’s disease. Med. Hypotheses. 2006;66(3):501–503. doi: 10.1016/j.mehy.2005.09.031. [DOI] [PubMed] [Google Scholar]
  • 19.O’Brien R.J., Wong P.C. Amyloid precursor protein processing and Alzheimer’s disease. Annu. Rev. Neurosci. 2011;34:185–204. doi: 10.1146/annurev-neuro-061010-113613. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Manev H. 5-Lipoxygenase gene polymorphism and onset of Alzheimer’s disease. Med. Hypotheses. 2000;54(1):75–76. doi: 10.1054/mehy.1998.0824. [DOI] [PubMed] [Google Scholar]
  • 21.Almeida Z.L., Brito R.M.M. Structure and aggregation mechanisms in amyloids.S. Molecules. 2020;25(5):1195. doi: 10.3390/molecules25051195. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Butterfield D.A., Swomley A.M., Sultana R. Amyloid β-peptide (1-42)-induced oxidative stress in Alzheimer disease: importance in disease pathogenesis and progression. Antioxid. Redox Signal. 2013;19(8):823–835. doi: 10.1089/ars.2012.5027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Liao Y-F., Wang B-J., Cheng H-T., Kuo L-H., Wolfe M.S. Tumor necrosis factor-, interleukin-1, and interferon-stimulate-secretasemediated cleavage of amyloid precursor protein through a JNK-dependent MAPK pathway. J. Biol. Chem. 2004;279(47):49523–49532. doi: 10.1074/jbc.M402034200. [DOI] [PubMed] [Google Scholar]
  • 24.Murphy M.P., LeVine H., III Alzheimer’s disease and the amyloid-β peptide. J. Alzheimers Dis. 2010;19(1):311–323. doi: 10.3233/JAD-2010-1221. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Sadigh-Eteghad S., Sabermarouf B., Majdi A., Talebi M., Farhoudi M., Mahmoudi J. Amyloid-beta: a crucial factor in Alzheimer’s disease. Med. Princ. Pract. 2015;24(1):1–10. doi: 10.1159/000369101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Stockley J.H., O’Neill C. Understanding BACE1: essential protease for amyloid-β production in Alzheimer’s disease. Cell. Mol. Life Sci. 2008;65(20):3265–3289. doi: 10.1007/s00018-008-8271-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Iqbal K., Liu F., Gong C-X., Alonso Adel.C., Grundke-Iqbal I. Mechanisms of tau-induced neurodegeneration. Acta Neuropathol. 2009;118(1):53–69. doi: 10.1007/s00401-009-0486-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Wang J.Z., Grundke-Iqbal I., Iqbal K. Kinases and phosphatases and tau sites involved in Alzheimer neurofibrillary degeneration. Eur. J. Neurosci. 2007;25(1):59–68. doi: 10.1111/j.1460-9568.2006.05226.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Bloom G.S. Amyloid-β and tau: the trigger and bullet in Alzheimer disease pathogenesis. JAMA Neurol. 2014;71(4):505–508. doi: 10.1001/jamaneurol.2013.5847. [DOI] [PubMed] [Google Scholar]
  • 30.van der Kant R., Goldstein L.S., Ossenkoppele R. Amyloid-β-independent regulators of tau pathology in Alzheimer disease. Nat. Rev. Neurosci. 2020;21(1):21–35. doi: 10.1038/s41583-019-0240-3. [DOI] [PubMed] [Google Scholar]
  • 31.Guo T., Noble W., Hanger D.P. Roles of tau protein in health and disease. Acta Neuropathol. 2017;133(5):665–704. doi: 10.1007/s00401-017-1707-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Alonso A.D., Grundke-Iqbal I., Barra H.S., Iqbal K. Abnormal phosphorylation of tau and the mechanism of Alzheimer neurofibrillary degeneration: sequestration of microtubule-associated proteins 1 and 2 and the disassembly of microtubules by the abnormal tau. Proc. Natl. Acad. Sci. USA. 1997;94(1):298–303. doi: 10.1073/pnas.94.1.298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Chang H.Y., Sang T-K., Chiang A-S. Untangling the Tauopathy for Alzheimer’s disease and parkinsonism. J. Biomed. Sci. 2018;25(1):54. doi: 10.1186/s12929-018-0457-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Köpke E., Tung Y-C., Shaikh S., Alonso A.C., Iqbal K., Grundke-Iqbal I. Microtubule-associated protein tau. Abnormal phosphorylation of a non-paired helical filament pool in Alzheimer disease. J. Biol. Chem. 1993;268(32):24374–24384. [PubMed] [Google Scholar]
  • 35.Lippens G., Sillen A., Landrieu I., Amniai L., Sibille N., Barbier P., Leroy A., Hanoulle X., Wieruszeski J-M. Tau aggregation in Alzheimer’s disease: what role for phosphorylation? Prion. 2007;1(1):21–25. doi: 10.4161/pri.1.1.4055. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Šimić G., Babić Leko M., Wray S., Harrington C., Delalle I., Jovanov-Milošević N., Bažadona D., Buée L., de Silva R., Di Giovanni G., Wischik C., Hof P.R. Tau protein hyperphosphorylation and aggregation in Alzheimer’s disease and other tauopathies, and possible neuroprotective strategies. Biomolecules. 2016;6(1):6. doi: 10.3390/biom6010006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Cummings J., Aisen P.S., DuBois B., Frölich L., Jack C.R., Jr, Jones R.W., Morris J.C., Raskin J., Dowsett S.A., Scheltens P. Drug development in Alzheimer’s disease: the path to 2025. Alzheimers Res. Ther. 2016;8(1):39. doi: 10.1186/s13195-016-0207-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Hardy J., Selkoe D. J. The amyloid hypothesis of Alzheimer's disease: progress and problems on the road to therapeutics. Science. 2002;297(5580):353–356. doi: 10.1126/science.1072994. [DOI] [PubMed] [Google Scholar]
  • 39.Morris G.P., Clark I.A., Vissel B. Inconsistencies and controversies surrounding the amyloid hypothesis of Alzheimer’s disease. Acta Neuropathol. Commun. 2014;2(1):135. doi: 10.1186/s40478-014-0135-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Guillozet A.L., Weintraub S., Mash D.C., Mesulam M.M. Neurofibrillary tangles, amyloid, and memory in aging and mild cognitive impairment. Arch. Neurol. 2003;60(5):729–736. doi: 10.1001/archneur.60.5.729. [DOI] [PubMed] [Google Scholar]
  • 41.Nelson P.T., Alafuzoff I., Bigio E.H., Bouras C., Braak H., Cairns N.J., Castellani R.J., Crain B.J., Davies P., Del Tredici K., Duyckaerts C., Frosch M.P., Haroutunian V., Hof P.R., Hulette C.M., Hyman B.T., Iwatsubo T., Jellinger K.A., Jicha G.A., Kövari E., Kukull W.A., Leverenz J.B., Love S., Mackenzie I.R., Mann D.M., Masliah E., McKee A.C., Montine T.J., Morris J.C., Schneider J.A., Sonnen J.A., Thal D.R., Trojanowski J.Q., Troncoso J.C., Wisniewski T., Woltjer R.L., Beach T.G. Correlation of Alzheimer disease neuropathologic changes with cognitive status: a review of the literature. J. Neuropathol. Exp. Neurol. 2012;71(5):362–381. doi: 10.1097/NEN.0b013e31825018f7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Nelson P.T., Braak H., Markesbery W.R. Neuropathology and cognitive impairment in Alzheimer disease: a complex but coherent relationship. J. Neuropathol. Exp. Neurol. 2009;68(1):1–14. doi: 10.1097/NEN.0b013e3181919a48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Akama K.T., Van Eldik L.J. β-amyloid stimulation of inducible nitric-oxide synthase in astrocytes is interleukin-1β- and tumor necrosis factor-α (TNFalpha)-dependent, and involves a TNFalpha receptor-associated factor- and NFkappaB-inducing kinase-dependent signaling mechanism. J. Biol. Chem. 2000;275(11):7918–7924. doi: 10.1074/jbc.275.11.7918. [DOI] [PubMed] [Google Scholar]
  • 44.Bennett D.A., Schneider J.A., Wilson R.S., Bienias J.L., Arnold S.E. Neurofibrillary tangles mediate the association of amyloid load with clinical Alzheimer disease and level of cognitive function. Arch. Neurol. 2004;61(3):378–384. doi: 10.1001/archneur.61.3.378. [DOI] [PubMed] [Google Scholar]
  • 45.Furcila D., Domínguez-Álvaro M., DeFelipe J., Alonso-Nanclares L. Subregional density of neurons, neurofibrillary tangles and amyloid plaques in the hippocampus of patients with Alzheimer’s disease. Front. Neuroanat. 2019;13:99. doi: 10.3389/fnana.2019.00099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Cribbs D.H., Berchtold N.C., Perreau V., Coleman P.D., Rogers J., Tenner A.J., Cotman C.W. Extensive innate immune gene activation accompanies brain aging, increasing vulnerability to cognitive decline and neurodegeneration: a microarray study. J. Neuroinflammation. 2012;9(1):179. doi: 10.1186/1742-2094-9-179. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Zimmer E.R., Leuzy A., Benedet A.L., Breitner J., Gauthier S., Rosa-Neto P. Tracking neuroinflammation in Alzheimer’s disease: the role of positron emission tomography imaging. J. Neuroinflammation. 2014;11(1):120. doi: 10.1186/1742-2094-11-120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Ferreira S.T., Clarke J.R., Bomfim T.R., De Felice F.G. Inflammation, defective insulin signaling, and neuronal dysfunction in Alzheimer’s disease. Alzheimers Dement. 2014;10(1) Suppl.:S76–S83. doi: 10.1016/j.jalz.2013.12.010. [DOI] [PubMed] [Google Scholar]
  • 49.Grammas P. Neurovascular dysfunction, inflammation and endothelial activation: implications for the pathogenesis of Alzheimer’s disease. J. Neuroinflammation. 2011;8(1):26. doi: 10.1186/1742-2094-8-26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.MERAZ RIOS M. A.; Toral-Rios, D.; Franco-Bocanegra, D.; Villeda-Hernández, J.; Campos-Peña, V., Inflammatory process in Alzheimer’s Disease. Front. Integr. Nuerosci. 2013;7:59. doi: 10.3389/fnint.2013.00059. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Azizi G., Navabi S.S., Al-Shukaili A., Seyedzadeh M.H., Yazdani R., Mirshafiey A. The role of inflammatory mediators in the pathogenesis of Alzheimer’s disease. Sultan Qaboos Univ. Med. J. 2015;15(3):e305–e316. doi: 10.18295/squmj.2015.15.03.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Das Sarma J. Microglia-mediated neuroinflammation is an amplifier of virus-induced neuropathology. J. Neurovirol. 2014;20(2):122–136. doi: 10.1007/s13365-013-0188-4. [DOI] [PubMed] [Google Scholar]
  • 53.Gomez-Nicola D., Perry V.H. Microglial dynamics and role in the healthy and diseased brain: a paradigm of functional plasticity. Neuroscientist. 2015;21(2):169–184. doi: 10.1177/1073858414530512. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Kettenmann H., Hanisch U.K., Noda M., Verkhratsky A. Physiology of microglia. Physiol. Rev. 2011;91(2):461–553. doi: 10.1152/physrev.00011.2010. [DOI] [PubMed] [Google Scholar]
  • 55.Schiweck J., Eickholt B.J., Murk K. Important shapeshifter: mechanisms allowing astrocytes to respond to the changing nervous system during development, injury and disease. Front. Cell. Neurosci. 2018;12:261. doi: 10.3389/fncel.2018.00261. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Eyo U.B., Dailey M.E. Microglia: key elements in neural development, plasticity, and pathology. J. Neuroimmune Pharmacol. 2013;8(3):494–509. doi: 10.1007/s11481-013-9434-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Nolte C., Möller T., Walter T., Kettenmann H. Complement 5a controls motility of murine microglial cells in vitro via activation of an inhibitory G-protein and the rearrangement of the actin cytoskeleton. Neuroscience. 1996;73(4):1091–1107. doi: 10.1016/0306-4522(96)00106-6. [DOI] [PubMed] [Google Scholar]
  • 58.Fu R., Shen Q., Xu P., Luo J.J., Tang Y. Phagocytosis of microglia in the central nervous system diseases. Mol. Neurobiol. 2014;49(3):1422–1434. doi: 10.1007/s12035-013-8620-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Bolmont T., Haiss F., Eicke D., Radde R., Mathis C.A., Klunk W.E., Kohsaka S., Jucker M., Calhoun M.E. Dynamics of the microglial/amyloid interaction indicate a role in plaque maintenance. J. Neurosci. 2008;28(16):4283–4292. doi: 10.1523/JNEUROSCI.4814-07.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Baik S.H., Kang S., Son S.M., Mook-Jung I. Microglia contributes to plaque growth by cell death due to uptake of amyloid β in the brain of Alzheimer’s disease mouse model. Glia. 2016;64(12):2274–2290. doi: 10.1002/glia.23074. [DOI] [PubMed] [Google Scholar]
  • 61.Stalder M., Phinney A., Probst A., Sommer B., Staufenbiel M., Jucker M. Association of microglia with amyloid plaques in brains of APP23 transgenic mice. Am. J. Pathol. 1999;154(6):1673–1684. doi: 10.1016/S0002-9440(10)65423-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Ries M., Sastre M. Mechanisms of Aβ clearance and degradation by glial cells. Front. Aging Neurosci. 2016;8:160. doi: 10.3389/fnagi.2016.00160. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Michelucci A., Heurtaux T., Grandbarbe L., Morga E., Heuschling P. Characterization of the microglial phenotype under specific pro-inflammatory and anti-inflammatory conditions: Effects of oligomeric and fibrillar amyloid-β. J. Neuroimmunol. 2009;210(1-2):3–12. doi: 10.1016/j.jneuroim.2009.02.003. [DOI] [PubMed] [Google Scholar]
  • 64.Wyss-Coray T., Loike J.D., Brionne T.C., Lu E., Anankov R., Yan F., Silverstein S.C., Husemann J. Adult mouse astrocytes degrade amyloid-β in vitro and in situ. Nat. Med. 2003;9(4):453–457. doi: 10.1038/nm838. [DOI] [PubMed] [Google Scholar]
  • 65.Rossner S., Lange-Dohna C., Zeitschel U., Perez-Polo J.R. Alzheimer’s disease β-secretase BACE1 is not a neuron-specific enzyme. J. Neurochem. 2005;92(2):226–234. doi: 10.1111/j.1471-4159.2004.02857.x. [DOI] [PubMed] [Google Scholar]
  • 66.Aguirre-Rueda D., Guerra-Ojeda S., Aldasoro M., Iradi A., Obrador E., Mauricio M.D., Vila J.M., Marchio P., Valles S.L. WIN 55,212-2, agonist of cannabinoid receptors, prevents amyloid β1-42 effects on astrocytes in primary culture. PLoS One. 2015;10(4):e0122843. doi: 10.1371/journal.pone.0122843. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Rådmark O., Samuelsson B. 5-Lipoxygenase: mechanisms of regulation. J. Lipid Res. 2009;50(Suppl.):S40–S45. doi: 10.1194/jlr.R800062-JLR200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Rådmark O., Werz O., Steinhilber D., Samuelsson B. 5-Lipoxygenase: regulation of expression and enzyme activity. Trends Biochem. Sci. 2007;32(7):332–341. doi: 10.1016/j.tibs.2007.06.002. [DOI] [PubMed] [Google Scholar]
  • 69.Ochs M.J., Suess B., Steinhilber D. 5-lipoxygenase mRNA and protein isoforms. Basic Clin. Pharmacol. Toxicol. 2014;114(1):78–82. doi: 10.1111/bcpt.12115. [DOI] [PubMed] [Google Scholar]
  • 70.Gaschler M.M., Stockwell B.R. Lipid peroxidation in cell death. Biochem. Biophys. Res. Commun. 2017;482(3):419–425. doi: 10.1016/j.bbrc.2016.10.086. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Higdon A., Diers A.R., Oh J.Y., Landar A., Darley-Usmar V.M. Cell signalling by reactive lipid species: new concepts and molecular mechanisms. Biochem. J. 2012;442(3):453–464. doi: 10.1042/BJ20111752. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Lewis R.A., Austen K.F., Soberman R.J. Leukotrienes and other products of the 5-lipoxygenase pathway. Biochemistry and relation to pathobiology in human diseases. N. Engl. J. Med. 1990;323(10):645–655. doi: 10.1056/NEJM199009063231006. [DOI] [PubMed] [Google Scholar]
  • 73.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(12):943–951. doi: 10.1111/cns.12596. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.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(3):768–778. doi: 10.1124/jpet.111.186031. [DOI] [PubMed] [Google Scholar]
  • 75.Di Gennaro A., Carnini C., Buccellati C., Ballerio R., Zarini S., Fumagalli F., Viappiani S., Librizzi L., Hernandez A., Murphy R.C., Constantin G., De Curtis M., Folco G., Sala A. Cysteinyl-leukotrienes receptor activation in brain inflammatory reactions and cerebral edema formation: a role for transcellular biosynthesis of cysteinyl-leukotrienes. FASEB J. 2004;18(7):842–844. doi: 10.1096/fj.03-0599fje. [DOI] [PubMed] [Google Scholar]
  • 76.Michael J., Marschallinger J., Aigner L. The leukotriene signaling pathway: a druggable target in Alzheimer’s disease. Drug Discov. Today. 2019;24(2):505–516. doi: 10.1016/j.drudis.2018.09.008. [DOI] [PubMed] [Google Scholar]
  • 77.Rouzer C.A., Matsumoto T., Samuelsson B. Single protein from human leukocytes possesses 5-lipoxygenase and leukotriene A4 synthase activities. Proc. Natl. Acad. Sci. USA. 1986;83(4):857–861. doi: 10.1073/pnas.83.4.857. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Shimizu T., Rådmark O., Samuelsson B. Enzyme with dual lipoxygenase activities catalyzes leukotriene A4 synthesis from arachidonic acid. Proc. Natl. Acad. Sci. USA. 1984;81(3):689–693. doi: 10.1073/pnas.81.3.689. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Jo-Watanabe A., Okuno T., Yokomizo T. The role of leukotrienes as potential therapeutic targets in allergic disorders. Int. J. Mol. Sci. 2019;20(14):3580. doi: 10.3390/ijms20143580. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Kanaoka Y., Boyce J.A. Cysteinyl leukotrienes and their receptors; emerging concepts. Allergy Asthma Immunol. Res. 2014;6(4):288–295. doi: 10.4168/aair.2014.6.4.288. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Lammers C.H., Schweitzer P., Facchinetti P., Arrang J.M., Madamba S.G., Siggins G.R., Piomelli D. Arachidonate 5-lipoxygenase and its activating protein: prominent hippocampal expression and role in somatostatin signaling. J. Neurochem. 1996;66(1):147–152. doi: 10.1046/j.1471-4159.1996.66010147.x. [DOI] [PubMed] [Google Scholar]
  • 82.Halliday G. Pathology and hippocampal atrophy in Alzheimer’s disease. Lancet Neurol. 2017;16(11):862–864. doi: 10.1016/S1474-4422(17)30343-5. [DOI] [PubMed] [Google Scholar]
  • 83.Jiang Z., Yin X., Jiang Q. Natural forms of vitamin E and 13′- carboxychromanol, a long-chain vitamin E metabolite, inhibit leukotriene generation from stimulated neutrophils by blocking calcium influx and suppressing 5-lipoxygenase activity, respectively. J. Immunol. 2011;186(2):1173–1179. doi: 10.4049/jimmunol.1002342. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Gilbert N.C., Gerstmeier J., Schexnaydre E.E., Börner F., Garscha U., Neau D.B., Werz O., Newcomer M.E. Structural and mechanistic insights into 5-lipoxygenase inhibition by natural products. Nat. Chem. Biol. 2020;16(7):783–790. doi: 10.1038/s41589-020-0544-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Chu J., Praticò D. Involvement of 5-lipoxygenase activating protein in the amyloidotic phenotype of an Alzheimer’s disease mouse model. J. Neuroinflammation. 2012;9(1):127. doi: 10.1186/1742-2094-9-127. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Zhang R-L., Lu C-Z., Ren H-M., Xiao B-G. Metabolic changes of arachidonic acid after cerebral ischemia-reperfusion in diabetic rats. Exp. Neurol. 2003;184(2):746–752. doi: 10.1016/S0014-4886(03)00296-6. [DOI] [PubMed] [Google Scholar]
  • 87.Liang G., Shi B., Luo W., Yang J. The protective effect of caffeic acid on global cerebral ischemia-reperfusion injury in rats. Behav. Brain Funct. 2015;11(1):18. doi: 10.1186/s12993-015-0064-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Firuzi O., Zhuo J., Chinnici C.M., Wisniewski T., Praticò D. 5-Lipoxygenase gene disruption reduces amyloid-β pathology in a mouse model of Alzheimer’s disease. FASEB J. 2008;22(4):1169–1178. doi: 10.1096/fj.07-9131.com. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Ikonomovic M.D., Abrahamson E.E., Uz T., Manev H., Dekosky S.T. Increased 5-lipoxygenase immunoreactivity in the hippocampus of patients with Alzheimer’s disease. J. Histochem. Cytochem. 2008;56(12):1065–1073. doi: 10.1369/jhc.2008.951855. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Czapski G.A., Czubowicz K., Strosznajder J.B., Strosznajder R.P. The lipoxygenases: their regulation and implication in Alzheimer’s disease. Neurochem. Res. 2016;41(1-2):243–257. doi: 10.1007/s11064-015-1776-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.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(3):590–599. doi: 10.1016/j.neurobiolaging.2013.09.036. [DOI] [PubMed] [Google Scholar]
  • 92.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. doi: 10.1016/j.neuropharm.2014.01.011. [DOI] [PubMed] [Google Scholar]
  • 93.Tang S-S., Ji M.J., Chen L., Hu M., Long Y., Li Y.Q., Miao M.X., Li J.C., Li N., Ji H., Chen X.J., Hong H. Protective effect of pranlukast on Aβ₁₋₄₂-induced cognitive deficits associated with downregulation of cysteinyl leukotriene receptor 1. Int. J. Neuropsychopharmacol. 2014;17(4):581–592. doi: 10.1017/S1461145713001314. [DOI] [PubMed] [Google Scholar]
  • 94.Voelkel N.F., Peters-Golden M. A new treatment for severe pulmonary arterial hypertension based on an old idea: inhibition of 5-lipoxygenase. Pulm. Circ. 2020;10(1):2045894019882635. doi: 10.1177/2045894019882635. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Liu Q., Zhao B. Nicotine attenuates β-amyloid peptide-induced neurotoxicity, free radical and calcium accumulation in hippocampal neuronal cultures. Br. J. Pharmacol. 2004;141(4):746–754. doi: 10.1038/sj.bjp.0705653. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Lewerenz J., Maher P. Chronic glutamate toxicity in neurodegenerative diseases—what is the evidence? Front. Neurosci. 2015;9:469. doi: 10.3389/fnins.2015.00469. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Wang Z-J., Zhou B., Mao W-W., Yin M. Overexpression of 5-lipoxygenase increases the neuronal vulnerability of PC12 cells to Aβ₄₂. Yakugaku Zasshi. 2011;131(12):1843–1853. doi: 10.1248/yakushi.131.1843. [DOI] [PubMed] [Google Scholar]
  • 98.Valera E., Dargusch R., Maher P.A., Schubert D. Modulation of 5-lipoxygenase in proteotoxicity and Alzheimer’s disease. J. Neurosci. 2013;33(25):10512–10525. doi: 10.1523/JNEUROSCI.5183-12.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Corser-Jensen C.E., Goodell D.J., Freund R.K., Serbedzija P., Murphy R.C., Farias S.E., Dell’Acqua M.L., Frey L.C., Serkova N., Heidenreich K.A. Blocking leukotriene synthesis attenuates the pathophysiology of traumatic brain injury and associated cognitive deficits. Exp. Neurol. 2014;256:7–16. doi: 10.1016/j.expneurol.2014.03.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Funk C.D. Leukotriene modifiers as potential therapeutics for cardiovascular disease. Nat. Rev. Drug Discov. 2005;4(8):664–672. doi: 10.1038/nrd1796. [DOI] [PubMed] [Google Scholar]
  • 101.Ström J.O., Strid T., Hammarström S. Disruption of the alox5ap gene ameliorates focal ischemic stroke: possible consequence of impaired leukotriene biosynthesis. BMC Neurosci. 2012;13(1):146. doi: 10.1186/1471-2202-13-146. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Giannopoulos P.F., Chu J., Joshi Y.B., Sperow M., Li J-G., Kirby L.G., Praticò D. 5-lipoxygenase activating protein reduction ameliorates cognitive deficit, synaptic dysfunction, and neuropathology in a mouse model of Alzheimer’s disease. Biol. Psychiatry. 2013;74(5):348–356. doi: 10.1016/j.biopsych.2013.04.009. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 103.Joshi Y.B., Chu J., Praticò D. Knockout of 5-lipoxygenase prevents dexamethasone-induced tau pathology in 3xTg mice. Aging Cell. 2013;12(4):706–711. doi: 10.1111/acel.12096. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Ivanov I., Heydeck D., Hofheinz K., Roffeis J., O’Donnell V.B., Kuhn H., Walther M. Molecular enzymology of lipoxygenases. Arch. Biochem. Biophys. 2010;503(2):161–174. doi: 10.1016/j.abb.2010.08.016. [DOI] [PubMed] [Google Scholar]
  • 105.Kuhn H., Banthiya S., Van Leyen K. Mammalian lipoxygenases and their biological relevance. Biochimica et Biophysica Acta (BBA)-. Molecular and Cell Biology of Lipids. 2015;1851(4):308–330. doi: 10.1016/j.bbalip.2014.10.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Skrzypczak-Jankun E., Jankun J., Al-Senaidy A. Human lipoxygenase: developments in its structure, function, relevance to diseases and challenges in drug development. Curr. Med. Chem. 2012;19(30):5122–5127. doi: 10.2174/092986712803530520. [DOI] [PubMed] [Google Scholar]
  • 107.Natarajan R., Nadler J.L. Lipid inflammatory mediators in diabetic vascular disease. Arterioscler. Thromb. Vasc. Biol. 2004;24(9):1542–1548. doi: 10.1161/01.ATV.0000133606.69732.4c. [DOI] [PubMed] [Google Scholar]
  • 108.Haeggström J.Z., Funk C.D. Lipoxygenase and leukotriene pathways: biochemistry, biology, and roles in disease. Chem. Rev. 2011;111(10):5866–5898. doi: 10.1021/cr200246d. [DOI] [PubMed] [Google Scholar]
  • 109.Elias I., Ferré T., Vilà L., Muñoz S., Casellas A., Garcia M., Molas M., Agudo J., Roca C., Ruberte J., Bosch F., Franckhauser S. ALOX5AP overexpression in adipose tissue leads to LXA4 production and protection against diet-induced obesity and insulin resistance. Diabetes. 2016;65(8):2139–2150. doi: 10.2337/db16-0040. [DOI] [PubMed] [Google Scholar]
  • 110.Liu Y-J., Fan H-B., Jin Y., Ren C-G., Jia X-E., Wang L., Chen Y., Dong M., Zhu K-Y., Dong Z-W., Ye B.X., Zhong Z., Deng M., Liu T.X., Ren R. Cannabinoid receptor 2 suppresses leukocyte inflammatory migration by modulating the JNK/c-Jun/Alox5 pathway. J. Biol. Chem. 2013;288(19):13551–13562. doi: 10.1074/jbc.M113.453811. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Joshi Y.B., Praticò D. The 5-lipoxygenase pathway: oxidative and inflammatory contributions to the Alzheimer’s disease phenotype. Front. Cell. Neurosci. 2015;8:436. doi: 10.3389/fncel.2014.00436. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Joshi Y.B., Praticò D. Knockout of 5-lipoxygenase results in age-dependent anxiety-like behavior in female mice. PLoS One. 2011;6(12):e29448. doi: 10.1371/journal.pone.0029448. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Chu J., Li J.G., Praticò D. Zileuton improves memory deficits, amyloid and tau pathology in a mouse model of Alzheimer’s disease with plaques and tangles. PLoS One. 2013;8(8):e70991. doi: 10.1371/journal.pone.0070991. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Praticò D., Zhukareva V., Yao Y., Uryu K., Funk C.D., Lawson J.A., Trojanowski J.Q., Lee V.M. 12/15-lipoxygenase is increased in Alzheimer’s disease: possible involvement in brain oxidative stress. Am. J. Pathol. 2004;164(5):1655–1662. doi: 10.1016/S0002-9440(10)63724-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Cole B.K., Lieb D.C., Dobrian A.D., Nadler J.L. 12- and 15-lipoxygenases in adipose tissue inflammation. Prostaglandins Other Lipid Mediat. 2013;104-105:84–92. doi: 10.1016/j.prostaglandins.2012.07.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Chinnici C.M., Yao Y., Ding T., Funk C.D., Praticò D. Absence of 12/15 lipoxygenase reduces brain oxidative stress in apolipoprotein E-deficient mice. Am. J. Pathol. 2005;167(5):1371–1377. doi: 10.1016/S0002-9440(10)61224-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Steinhilber D., Hofmann B. Recent advances in the search for novel 5-lipoxygenase inhibitors. Basic Clin. Pharmacol. Toxicol. 2014;114(1):70–77. doi: 10.1111/bcpt.12114. [DOI] [PubMed] [Google Scholar]
  • 118.Leyen K. v. Lipoxygenase: an emerging target for stroke therapy. CNS & Neurological Disorders-Drug Targets (Formerly Current Drug Targets-CNS & Neurological Disorders) . 2013;12(2):191–199. doi: 10.2174/18715273112119990053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Bain G., King C.D., Brittain J., Hartung J.P., Dearmond I., Stearns B., Truong Y.P., Hutchinson J.H., Evans J.F., Holme K. Pharmacodynamics, pharmacokinetics, and safety of AM211: a novel and potent antagonist of the prostaglandin D2 receptor type 2. J. Clin. Pharmacol. 2012;52(10):1482–1493. doi: 10.1177/0091270011421912. [DOI] [PubMed] [Google Scholar]
  • 120.Follows R.M., Snowise N.G., Ho S-Y., Ambery C.L., Smart K., McQuade B.A. Efficacy, safety and tolerability of GSK2190915, a 5-lipoxygenase activating protein inhibitor, in adults and adolescents with persistent asthma: a randomised dose-ranging study. Respir. Res. 2013;14(1):54. doi: 10.1186/1465-9921-14-54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Gür Z.T., Çalışkan B., Banoglu E. Drug discovery approaches targeting 5-lipoxygenase-activating protein (FLAP) for inhibition of cellular leukotriene biosynthesis. Eur. J. Med. Chem. 2018;153:34–48. doi: 10.1016/j.ejmech.2017.07.019. [DOI] [PubMed] [Google Scholar]
  • 122.Halle A., Hornung V., Petzold G.C., Stewart C.R., Monks B.G., Reinheckel T., Fitzgerald K.A., Latz E., Moore K.J., Golenbock D.T. The NALP3 inflammasome is involved in the innate immune response to amyloid-β. Nat. Immunol. 2008;9(8):857–865. doi: 10.1038/ni.1636. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Heneka M.T., Kummer M.P., Stutz A., Delekate A., Schwartz S., Vieira-Saecker A., Griep A., Axt D., Remus A., Tzeng T-C., Gelpi E., Halle A., Korte M., Latz E., Golenbock D.T. NLRP3 is activated in Alzheimer’s disease and contributes to pathology in APP/PS1 mice. Nature. 2013;493(7434):674–678. doi: 10.1038/nature11729. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Lewis J., Dickson D.W. Propagation of tau pathology: hypotheses, discoveries, and yet unresolved questions from experimental and human brain studies. Acta Neuropathol. 2016;131(1):27–48. doi: 10.1007/s00401-015-1507-z. [DOI] [PubMed] [Google Scholar]

Articles from Current Neuropharmacology are provided here courtesy of Bentham Science Publishers

RESOURCES