You are on page 1of 11

Biomedicine & Pharmacotherapy 144 (2021) 112250

Contents lists available at ScienceDirect

Biomedicine & Pharmacotherapy


journal homepage: www.elsevier.com/locate/biopha

Review

Mechanistic role of boswellic acids in Alzheimer’s disease: Emphasis on


anti-inflammatory properties
Aisha Siddiqui a, Zahoor Shah b, Rao Nargis Jahan c, Iekhsan Othman d, Yatinesh Kumari a, *
a
Neurological disorder and aging research group (NDA), Microbiome and Bioresource Research Strength (MBRS), Jeffrey Cheah School of Medicine and Health Sciences,
Monash University Malaysia, 47500, Selangor, Malaysia
b
Department of Medicinal and Biological Chemistry, University of Toledo, 3000 Arlington Avenue, Toledo 43614, OH, USA
c
Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard University, New Delhi 110062, India
d
Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500 Selangor, Malaysia

A R T I C L E I N F O A B S T R A C T

Keywords: The resin/gum of Boswellia species belonging to the family of Burseraceae is a naturally occurring mixture of
Boswellic acids bioactive compounds, which was traditionally used as a folk medicine to treat conditions like chronic inflam­
Neuroinflammation mation. Several research studies have also explored its’ therapeutic potential against multiple neurodegenerative
Inflammatory mediators
diseases such as Alzheimer’s disease (AD). The main chemical constituents of this gum include boswellic acids
Alzheimer’s disease
Signalling pathways
(BAs) like 3-O-acetyl-11-keto-β boswellic acid (AKBA) that possess potent anti-inflammatory and neuroprotective
properties in AD. It is also involved in inhibiting the acetylcholinesterase (AChE) activity in the cholinergic
pathway and improve choline levels as well as its binding with nicotinic receptors to produce anti-inflammatory
effects. Multiple shreds of evidence have demonstrated that BAs modulate key molecular targets and signalling
pathways like 5-lipoxygenase/cyclooxygenase, Nrf2, NF-kB, cholinergic, amyloid-beta (Aβ), and neurofibrillary
tangles formation (NFTs) that are involved in AD progression. The present review focuses on the possible
mechanistic therapeutic role of BAs in modulating the 5-LOX/COX pathway in arachidonic acid metabolism,
activating Nrf2 through binding of ARE, inhibiting NF-kB and AChE activity. In addition, an inhibition of am­
yloid plaques (Aβ) and neurofibrillary tangles (NFTs) induced neurotoxicity and neuroinflammation in AD by
BAs is also discussed in this review. We have also highlighted that BAs possess beneficial effects in AD by tar­
geting multiple molecular pathways and makes it an emerging drug candidate for treating neurodegenerative
diseases.

1. Introduction molecules resulting in altered lipid metabolism [4]. In addition, previ­


ously formed senile plaques trigger elevated pro-inflammatory media­
Alzheimer’s disease (AD) is described as a neurological disorder tors like chemokines, cytokines, interleukins (ILs) that potentiate AD
primarily affecting the elderly population marked by dementia, progression. Moreover, the twin role of chemokine engagement as anti
impaired cognition, and changes in behaviour. According to Alzheimer’s and pro-inflammatory mediators is well understood, but clinically it
Disease International (2019) report it is estimated that approximately 50 needs to look upon further in AD progression [5,6]. These signalling
million of the population are suffering from dementia until the present molecules regulate central nervous system (CNS) functions and patho­
day, which may increase up to 152 million by 2050 [1]. genesis of neurological diseases like AD through different cascades that
Hallmarks of AD include Aβ senile plaque formation and neurofi­ contribute to inflammation- induced AD.
brillary tangle (NFTs) accumulation in the brain [2]. Soluble Aβ plaques The 1980 s report suggests that immune cells and proteins occupy the
develop shortly after NFTs formation that triggers defects in synaptic area close to β-amyloid plaques [7,8]. Moving to the 1990 s, many
functions, mitochondria dysfunction and increases the release of reac­ observational studies and epidemiological data revealed that the
tive oxygen species (ROS), contributing to oxidative stress [3]. More­ anti-inflammatory therapies for diseases like arthritis had shown ther­
over, additional signs of AD include excessive production of lipid apeutic efficacy against AD development. These patients were long-term

* Corresponding author.
E-mail address: Yatinesh.kumari@monash.edu (Y. Kumari).

https://doi.org/10.1016/j.biopha.2021.112250
Received 19 July 2021; Received in revised form 17 September 2021; Accepted 26 September 2021
Available online 1 October 2021
0753-3322/© 2021 The Authors. Published by Elsevier Masson SAS. This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/by-nc-nd/4.0/).
A. Siddiqui et al. Biomedicine & Pharmacotherapy 144 (2021) 112250

NSAIDS consumers and showed a 50% lower risk of AD development most popular ingredients used in alternative and complementary med­
[9–12]. These reports persuade to utilize transgenic AD animal models icine systems, which has shown therapeutic potential against gastroin­
to observe the mechanism of NSAIDS involved in AD pathologies [13]. testinal, inflammatory, microbial, and hormonal diseases [35].
Later, clinical trials using NSAIDS showed results but without objective Conventionally, the gum resin has also been used for cosmetic products,
evidence of effectiveness [14]. Such previous studies support the role of different adhesive, coating substances, the essence employed in tradi­
neuroinflammatory pathways involved in developing AD. Moreover, tional ceremonies.
neuroinflammation results from more than one risk factor associated Approximately 25 species of Boswellia are dispersed in the moun­
with AD pathologies that play to boost the disease severity by worsening tains of India, Arabia, and Northeast Africa. The resins in India are
tau and β-amyloid pathologies [15,16]. During inflammation NFTs and famous as salai guggul, which consists of volatile oils, mucus, and resin
Aβ plaques accumulate together with astrocytes, microglia, and other acids [35,36]. These resins are also comprised of penta and tetracyclic
immune cells in the brain [17]. Meanwhile, genome-wide association triterpenes. However, the percentage of the constituents differs from
studies (GWAS) have explained a strong correlation between immune species to species [Table 1] [37].
cells and AD development [18–20]. This investigation demonstrated
that Aβ plaque and tau formation are stimulated by pro-inflammatory 2.1. Penta and tetracyclic triterpenes
mediators [21,22]. During the chronic neuroinflammation, activated
immune cells like microglia release pro-inflammatory cytokines, ROS, Particularly 12 kinds of pentacyclic triterpenes have been identified
and nitric oxide and promote tau hyperphosphorylation, which con­ in Boswellia extract samples shown in [Table 2] [38]. Out of these
tributes to AD pathology [23]. However, activated immune cells samples, two are found to be most active i.e., 11-Keto-β-boswellic acid
through phagocytosis also stimulate the Aβ plaque clearance [24]. A (KBA) and Acetyl-11-keto-β-boswellic acid (AKBA).
study by Goldgaber et al. in 1989 had analysed the brain samples of dead There are three tetracyclic triterpenes which are 3-acetoxytirucallic
patients who suffered from a recent head injury and demonstrated Acid, 3-oxotirucallic acid and 3-hydroxytirucallic acid. Additional
elevated Aβ aggregates. It is evident that increased IL-1 expression is components that are pharmacologically active are: lupeolic acid, acetyl-
culpable for augmented amyloid precursor protein (APP) and Aβ pro­ lupeolic acid, betulinic acid, incensole acetate, incensole oxide, iso­
duction [25,26]. Improved IL-1β levels also increase other cytokines incensole oxide, isoincensole acetate [39].
production, in parallel with IL-6, which activates a cyclin dependant
kinase enzyme CDK5 for tau hyperphosphorylation [27]. In AD neuro­ 2.2. Pharmacokinetics of BAs
inflammation appears to function as a preliminary contributing factor in
exaggerating tau and Aβ load; however, it is still debatable whether Different boswellic acids (BAs) have shown therapeutic benefits
inflammation is a driving factor for AD or vice-versa [28]. against neurodegenerative diseases, and inflammatory conditions, it is
At present, only five FDA-approved drugs for AD are in use, including important to emphasize the pharmacokinetics of BAs, as these properties
4 cholinesterase inhibitors i.e., tacrine, donepezil, rivastigmine and play a crucial role in drug development to achieve maximal efficacy in
galantamine and one NMDA receptor antagonist memantine [29]. These vivo [41–43]. In this context, the concentration of different BAs (KBA,
drugs are more of a palliative therapy instead of a curative. [30]. AKBA, αBA, βBA, AαBA, AβBA) in rat brains were examined, and it has
However, in June 2021 the FDA approved monoclonal antibody, adu­ been found that after 8 hrs of oral administration of Boswellia serrata
canumab, an IgG1 anti–amyloid-β (Aβ) antibody has shown an emerging gum resin extract (240 mg/kg), the maximum levels of the above
scope against AD progression [31]. In this regard, a study conducted by mentioned BAs in the brain was found to be for βBA (1066.6 ng/g)
Jeff Sevigny and colleagues has demonstrated that aducanumab suc­ followed by αBA (485.1 ng/g), AβBA (163.7 ng/g), AαBA (43.0 ng/g),
cessfully cleared Aβ aggregates in a dose-dependent manner in the ro­ AKBA (37.5 ng/g) and KBA (11.6 ng/g) [44]. Furthermore, another
dent model of AD. Additionally, this therapeutic agent has been found to study analysed the concentrations of the two most potent BAs i.e., KBA
reduce Aβ plaques and cognitive decline in AD patients [32]. and AKBA in rat brains, and their level was found to be 99 and 95 ng/g of
The limited treatment options against AD, have prompted the re­ the brain, respectively. These concentrations of KBA and AKBA were
searchers to explore potent pharmacological agents/adjuvants with detected 3 h after peroral administration of 240 mg/kg of dry Boswellia
broad biological activity and fewer side effects to treat neurodegenera­ serrata to Wistar rats [45]. A recent study compared the concentration of
tive diseases. In view of this, bioactive compounds from natural sources AKBA in rat brains after administration through the intranasal and
have shown potential therapeutic benefits against various neurological sublingual route. It has been observed that AKBA has efficiently abled to
disorders, including AD [33]. One such biologically active compound is cross the blood brain barrier (BBB) and the bioavailability of AKBA was
boswellic acid (BA) which is procured from the oleo gum resin of the significantly higher in case of sublingual route when compared to
various Boswellia species. However, few species such as Boswellia ser­ intranasal administration, as demonstrated by area under curves (AUCs)
rata, Boswellia carteri, Boswellia sacra have been investigated to possess analysis [46]. A human pharmacokinetic study was performed to mea­
therapeutic potential for chronic diseases like cancer, arthritis, inflam­ sure the elimination half-life and plasma concentration of Boswellia
matory disease, Parkinson’s disease and Alzheimer’s disease. There is serrata extract (BSE). It has been noted that the elimination half-life of
still a broad scope to investigate the role of a bioactive component of the drug was about six hours and peak plasma levels of the drug reach 30
boswellic acids obtained from various Boswellia species [34]. In the hrs after oral administration of 333 mg of BSE. This study also suggested
present review, we discussed the individual role as well as the crosstalk that the drug needs to be administered at a dosing interval of 6 hrs.
of Nrf2, NF-kB cascades that majorly contribute to neuroinflammation, Moreover, the given dose was well tolerated by all the individuals and
neurotoxicity, and AD. Additionally, we have highlighted the patho­ no side effects were noted [47].
genesis of AD via the cholinergic pathway, Aβ and neurofibrillary tan­
gles formation that plays a crucial role in neuroinflammation,
neurotoxicity and apoptosis. Moreover, the role of BA in modulating
these pathways and molecular targets has also taken into consideration.
Table 1
Components of BA.
2. Boswellic Acids (BAs)
Boswellia serrata[37] Boswellia carteri Birdw[40]

Genus Boswellia belongs to the family Burseraceae, and it was tradi­ Volatile oils 7.5–9–15 5–9
tionally used as a medicine. Phytochemical constituents of this family Mucus ~23 ≈ 12–20%
Pure Resin 55–57 ~66
are pharmacologically active. Its gum/resin is one of the primary and

2
A. Siddiqui et al. Biomedicine & Pharmacotherapy 144 (2021) 112250

Table 2 COX2. However, the binding affinity of BAs to COX1 was significantly
Types of pentacyclic triterpenes in Boswellia extract. higher than COX2, indicating an increased sensitivity of COX1 for BAs as
Constituents Percentage noted in COX1 X-ray structures [69]. Another study revealed that BAs
possessed anti-inflammatory activity by inhibiting microsomal prosta­
α-Boswellic acid and β-Boswellic acid (β-BA) (α-BA) 10–21%[39]
Acetyl-α-boswellic acid (AαBA) and Acetyl-β-boswellic acid 0.05–6%[39] glandin E2 synthase-1 (mPGES1), thus preventing the conversion of
(AβBA) PGH2 to PGE2. [70]. In vivo study in rodents showed that BA inhibited
Acetyl-lupeolic acid and Lupeolic acid 1.10%, 2.61%[37, prostaglandin E2 synthase-1 for the anti-inflammatory activity [70].
38] AKBA, in combination with COX inhibitors, prevented oxidative
Acetyl-9–11-dehydro-α-boswellic acid and 11-Dehydro- 0.06%, 0.18%[37,
α-boswellic acid 38]
stress-induced neuronal damage and cognitive impairment because of
Acetyl-9–11-dehydro-β-boswellic acid and 9,11-Dehydro- 0.52%, 0.83%[37, the antioxidant, anti-inflammatory and anti-glutamatergic effects [62,
β-boswellic acid 38] 71]. In the LPS induced neuroinflammation rodent model, AKBA
11-Keto-β-boswellic acid (KBA) 2.5–7.5%[39] significantly reduced pro-inflammatory mediators such as 5-LOX,
Acetyl-11-keto-β-boswellic acid (AKBA) 0.1–4%[38,39]
TNF-α, IL-6 levels and improve cognition [72]. The pathway of 5-LOX/­
COX and the role of BA based on the above studies is described in Fig. 1.
3. Signaling pathways and molecular targets modulated by BAs
3.2. Nuclear factor erythroid 2–related factor 2 (Nrf2) signaling pathway
Several studies indicated that BA possesses pharmacological prop­
erties like antiseptic, antineurotic, anxiolytic, analgesic [48]. In addi­ Nrf2 is a member of cap ‘n’ collar family and is a chain of 605 amino
tion, it also showed potent anti-inflammatory activity against arthritis, acid which is in cytoplasm during the inactivated stage and upon acti­
asthma, cerebral edema, and cancer [49,50]. The modification in vation of various stimuli such as oxidative stress it translocates to the
transduction signaling pathways gives rise to previously mentioned nucleus to mediate its effects [73]. In basal conditions i.e., in cytoplasm
chronic conditions that might impact population of all ages [51,52]. In Nrf2 becomes the target of many ubiquitin molecules, whereas in the
such diseases, BAs exerts its therapeutic benefits by modulating various nucleus, redox homeostasis is regulated by Nrf2 accumulation, which
targets like growth factors, transcription factors, enzymes, kinases, and carries out the normal antioxidant response element (ARE) associated
receptors that may initiate apoptosis and finally arrest the cell cycle gene expression. Furthermore, Nrf2-ARE pathway in the cytoplasm is
[53]. It inhibits various pathways [54] associated with cell durability governed by separation and deterioration of Nrf2 through Kelch Like
[55], metastasis [56], proliferation [57]. ECH Associated Protein 1 (KEAP1) binding [74]. ARE activators or stress
The essential molecular mechanisms that have been investigated as stimuli make conformational changes in KEAP1 which unbind Nrf2 and
the biological targets of BAs are 5-Lipoxygenase (5-LOX), kinase enzyme allows it to translocate into the nucleus. Other processes like phos­
topoisomerase I, II, and Ikβ, human leukocyte elastase. Moreover, BAs phorylation by AMP-activated protein kinase (AMPK), acetylation, and
are known to regulate the Ca (2+/-) and mitogen-activated protein ki­ protein kinase C (PKC) also stimulate Nrf2 and KEAP1 separation [74].
nases (MAPK) mechanisms, it is also involved in the pathways that are Nrf2 is known for regulating the redox homeostasis process which
essential for inflammation and tumor progression [58–60]. involves activation of other pathways [75] consists of antioxidant and
anti-inflammatory molecules [76–80]. Nrf2 exerts its protective action
by regulating the function of molecules like heme oxygenase-1 (HO-1),
3.1. 5-Lipoxygenase (5-LOX) glutathione S-transferase (GST), superoxide dismutase (SOD), Nicotin­
amide adenine dinucleotide phosphate (NAD(P)H), malondialdehyde
The initial step of arachidonic acid cascade activation is to produce (MDA), brain-derived neurotrophic factor (BDNF), and prevents degra­
inflammatory mediators like IFN-γ (interferon-γ), TNFα (tumor necrosis dation of inhibitory kappa B alpha (IĸBα) thereby suppressing nuclear
factor α), ILs with the help of a membrane-linked enzyme phospholipase factor kappa β (NF-kB) dependent pro-inflammatory molecules expres­
A2 (PLA2). The two enzymes cyclooxygenase (COX-2) and 5-LOX are sion such as IL-6 [81]. As evident from a study in APPswe/PS1dE9 mice
responsible for the production of prostaglandins (PGs) and leukotrienes treated chronically with AKBA showed antioxidant properties by
(LTs), which finally precipitates to produce specific symptoms of improving the levels of SOD and GSH in the hippocampus and cerebral
inflammation [37]. Moreover, they are the components of arachidonic cortex regions when compared with the wild-type group. Similarly, such
acid oxygenation that show an increased expression in aging brain a treatment also resulted in a decreased expressions of TNF-α, IL-1β, IL-6
physiologies [61,62]. 5-LOX helps in the catalysis of arachidonic acid and MDA in both regions of the brain. These findings strongly suggest
into LTA4, which is further converted to Leukotriene B4 or C4 (LTB4, AKBA elicited Nrf2/HO-1/NF-kB-mediated antioxidant and
LTC4) [63]. When deposited in the brain, these leukotrienes play a sig­ anti-inflammatory responses [82].
nificant role in altering the pathology of brain tissues [64,65]. Many Downregulation of Nrf2 function increases the risk of cell damage by
research studies support the participation of 5-LOX in AD development. ROS and pro-inflammatory molecules [83–86]. The possible underlying
As evident from the comparative post-mortem study between healthy neuroprotective mechanism of Nrf2 could be attributed to its
and AD patient’s brains that expressed high levels of 5-LOX in the cortex anti-inflammatory activity, as demonstrated by the downregulated
and hippocampal region of AD brains. However, no considerable dif­ mRNA expression levels IL-6, inducible NO synthase, and TNF-α in mice
ference was observed in both healthy and AD brain’s cerebellum (an model of neuroinflammation [87]. Furthermore, ROS are known to
area devoid of AD pathogenesis) [66,67]. aggravate beta-site APP cleaving enzyme 1 (BACE1), which enhances
Furthermore, BA studies targeting inflammatory molecule 5-LOX the generation of Aβ plaques [88]. The presence of Aβ lesions represents
and COX enzymes came forward to explain the potential efficacy chronic inflammation in AD brains [89]. Data from the previous study
against inflammation cascades. One such recent study by Mortiz Verhoff on APP/PS1 mice suggest that disruption of Nrf2 cascades results in a
et. al; in 2014 suggested that penta and tetracyclic triterpene acid from considerable rise in APP, Aβ1–40 and Aβ1–42 [90]. Moreover, a study by
the wellknown Boswellia species has shown to inhibit prostaglandin Branca et al., explored that increase in amyloid plaques is faster in
synthesis by inhibiting the inflammatory action of COX enzymes [68]. APP/PS1/Nrf2− /− mice than APP/PS1 mice. However, the deficiency
Moreover, molecular docking studies have been helpful to understand of Nrf2 in APP/PS1 mice influences OS [91] and attenuation of Nrf2
the pharmacological interaction of ligands/drugs with the active sites of associated pathways in APP/PS1 mice occurs during Aβ aggregation.
the receptors. Further investigation utilizing an automated molecular Particularly, Nrf2 upregulation in in-vitro prevents Aβ induced neuro­
docking approach to elucidate the binding of BAs was performed and toxicity [92].
found a correlation between positive chemscore values for COX1 and This increased defense activity of Nrf2 against stimuli such as

3
A. Siddiqui et al. Biomedicine & Pharmacotherapy 144 (2021) 112250

Fig. 1. Represents the inhibition of 5–Lipoxygenase and cyclooxygenase by BA in AD pathogenesis. PLA2- phospholipase A2; IFN-γ-interferon-γ; IL-1/6; interleukin
1/6; 5-LOX- 5- Lipoxygenase; COX- cyclooxygenases; PGs- prostaglandins; Txs- thromboxanes; LTA4/B4/C4- leukotrienes; TNF-α- tumor necrosis factor α; BACE1-
beta-site APP cleaving enzyme 1; APP- amyloid precursor protein; Aβ- amyloid-beta; CDK5- cyclin dependant kinase enzyme; P- phosphorylation; AD- Alzheimer’s
disease; BA- boswellic acid.

oxidative stress and plaque accumulation occurs in response to inflam­ accelerate the recruitment and adhesion of immune cells at the in­
mation, injuries, and harmful substances. NF-κB also gets activated by flammatory site, NF- kB improves chemokine and adhesion molecules
similar stimuli; therefore, Nrf2- NF-κB together work as a team to expression [96]. In AD, NF-kB regulates the BACE1 and APP expression
equalize each other’s performance [74]. [97], leading to increased Aβ formation [98]. Another in vivo and in
vitro study showed that activated NF-kB effectuate β secretase levels in
3.3. Nuclear factor kappa-light-chain-enhancer of activated B cells (NF- neurons [99]. Recently, a study has demonstrated that AKBA at a dose of
kB) signaling pathway 5 mg/kg, when administered sublingually in a rodent model, has shown
a significant suppression of NF-kB signaling activity, as evident by
NF-kB pathway is characterized as a canonical and noncanonical decreased NF-kBp65 expression, to exert the neuroprotective effects.
pathway that gets activated as a response to various stimuli. Canonical These investigations substantiate that NF-kB cascades could be playing a
cascade involves activation of stimuli like cytokines after recognising significant role in AD development [46].
the receptors such as Toll-like receptors (TLRs), IL-1 receptor (IL-1R),
tumor necrosis factor receptor (TNFR) and antigen receptors that stim­ 3.4. Crosstalk between NRF2 and NF- ĸB
ulate signaling pathways eventually activating inhibitory nuclear factor
kappa β kinase (IKKβ). Activation of IKKβ leads to the phosphorylation of Alzheimer’s disease is strongly associated with neuroinflammation
IĸB proteins which establish the platform for ubiquitination and pro­ and neurodegeneration. Nrf2 and NF- kB are popularly determined as an
teosomal degeneration, therefore, relocating the NF-kB dimers into the essential component of inflammatory cascades and therefore indicating
nucleus, where binding to defined DNA sequences takes place via a promising approach for inflammation-derived AD therapy. These two
transcription of targeted genes to regulate oxidative stress, inflamma­ factors coordinate and balance oxidative stress and inflammation.
tion, and apoptosis. Noncanonical pathway includes selected constitu­ Various genetic and pharmacological research has shown important
ents of the cytokine group that is responsible for p100 phosphorylation crosstalk between Nrf2 and NF-kB pathways [100]. Nrf2 activation in­
and p52/RelB complex formation by activating inhibitory nuclear factor creases the expression of HO-1 and prevents IκB degradation, which
kappa α kinase (IKKα) [93,94]. inhibits NF-kB activation and ROS generation [101–103]. Likewise,
The underlying inflammatory, pro-inflammatory, growth response transcription mediated by NF-kB suppresses Nrf2 activation by
and stress genes are transcripted under NF- kB supervision [95]. Surely, decreasing ARE genes and unoccupied CREB binding protein (CBP) by
one of the important investigations of NF- kB role in aging and other fighting with Nrf2 for CBP thus suppressing Nrf2 activation [104,105].
pathophysiologies is the regulated transcription of cytokines levels like A recent study by Chao Wei et.al., has shown the neuroprotective
IL-1α/β, TNF-α and proteins like β2 microglobulin. Moreover, to role of AKBA through Nrf2 and NF-kB interplaying pathways. Chronic

4
A. Siddiqui et al. Biomedicine & Pharmacotherapy 144 (2021) 112250

administration of AKBA in APPswe/PS1dE9 mice brain has shown a cytotoxicity mechanism [115,116].
reduction in inflammatory mediators like TNF-α, IL-1β and IL-6. AKBA is Neurons after activation are dependent on choline for getting back to
presumed to selectively inhibit NK- kB pathway and activate Nrf2/HO1 their resting phase; therefore, choline is produced by the hydrolysis of
pathway. Moreover, AKBA treatment has shown improvement in syn­ ACh in the presence of AChE [117]. The cholinergic anti-inflammatory
aptic function through the recovery of synaptic integrity and plasticity, pathway involves the inhibition of TNF and other pro-inflammatory
ameliorating cognitive impairments due to reduced inflammation, cytokines release and control inflammation. This unique performance
oxidative stress and Aß production [82]. Ding et al. in 2014 has reported is forced by the efferent vagus nerve, which is evolved from the dorsal
that BAs has shown neuroprotection by significantly reducing infarction vagal nucleus within the medulla oblongata. Electrical stimulation of
volume and apoptosis in the Sprague Dawley rat model of vagus nerve (VNS) or guanyl hydrazone CNI-1493 communicates with
Oxygen-glucose deprived ischemic injury induced by oxidative stress. α7 subunit-consisting nicotinic receptors (α7nAchR). Activation of these
AKBA has shown enhanced Nrf2 binding to ARE. Therefore, this study nicotinic receptors involves cholinergic transmission that eventually
suggests that AKBA shows neuroprotective effects against ischaemic suppresses TNF synthesis and systemic inflammation [118]. Further­
injury via nuclear factor erythroid-2-related factor 2 (Nrf2)/heme more, α7nAchR stimulation on macrophages suppresses
oxygenase-1 (HO-1) cascade activation [106,107]. As discussed previ­ pro-inflammatory mediators, released by inhibiting NF-kB cascade
ously, the role of Nrf2 and NF-kB in AD and the therapeutic potential of [119]. Additionally, the administration of nicotine in epithelial cells
BA in modulating the above pathways are illustrated in Fig. 2. decreases TNF-α, IL-6, and IL-1β expression regulated by α7nAchR and
NF-kB signaling pathway [120]. Moreover, in the previous study it has
been observed that AChE also aggravates Aβ formation and neurotox­
3.5. Cholinergic anti-inflammatory pathway icity [121]. Therefore, many studies have been conducted to explore the
efficacy of BAs on the acetylcholinesterase pathway in AD. In a diseased
In AD, the decline in choline levels results in cortical dysfunction, state, the AChE activity was found to be increased, which results in
memory deficit, abnormal cerebral blood flow, task learning difficulty, decreased ACh levels leading to a loss in neuronal communication
disrupts the sleep cycle and cortex development [108]. The dysfunction [122]. Findings from a recent study suggested the reduced levels of ACh
in the acetyl choline pathway in AD takes place at different stages were improved as well as elevated activity of AChE was reduced in
involving reduced acetylcholinesterase (AChE) activity and choline chemo-preventive and chemo-therapeutic groups of BA in the rodent
uptake, less acetylcholine (ACh) synthesis and its receptors (AChR). In model of AD [123]. A possible pharmacodynamic activity that has been
AD, AChE plays a role independent of choline which contributes to in­ concluded yet is that BAs can significantly reduce/inhibit AChE in serum
flammatory responses [109–114], Aβ complex formation and

Fig. 2. Schematic representation of BA inhibiting the inflammatory mechanism in crosstalk between Nrf2 and NF-kB induced neurotoxicity and AD. ROS- reactive
oxygen species; Nrf2- Nuclear factor erythroid 2–related factor 2; Keap1- Kelch Like ECH Associated Protein 1; Ub- ubiquitination; AMPK- AMP-activated protein
kinase; PKC- protein kinase C; P- phosphorylation; A- acetylation; Ik-Bα-inhibitor of nuclear factor kappa B; NF-kB- Nuclear factor kappa-light-chain-enhancer of
activated B cells; ARE- antioxidant response element; HO-1- Haemeoxygenase 1; SOD- superoxide dismutase; GST- glutathione S-transferase; NADPH- nicotinamide
adenine dinucleotide phosphate; MDA malondialdehyde; BDNF- brain-derived neurotropic factor; BACE1- beta-site APP cleaving enzyme 1; APP- amyloid precursor
protein; Aβ- amyloid-beta; IL-1a/β - Interleukin 1a/β; IL-6- Interleukin 6; TNF-α- tumor necrosis factor α.

5
A. Siddiqui et al. Biomedicine & Pharmacotherapy 144 (2021) 112250

against trimethyltin (TMT) induced neurotoxicity in a rodent model to the accumulation of protein fragments which if not cleaned up by the
[124]. Since TMT is a neurotoxic agent that causes an increase in AChE lysosomal or autophagy pathway then accumulation begins leading to
levels leading to change in behaviour, learning and cognitive decline neurodegeneration [132]. Tau aggregation can also be initiated by
[125,126]. Moreover, increasing the dose of BA in the same model has polyanions trigger charge compensation of the middle portion of the tau
shown promising results in improving neuronal health of the hippo­ chain; therefore, hyperphosphorylated tau begins to form NFTs intra­
campus and cortex region when observed in the Morris water maze test cellularly which interferes in regular neuronal communication [133].
[124]. As illustrated below in Fig. 3 is the summary of the above finding. Moreover, in developed neurons, impaired axonal transport is generally
known to be a major factor for most of the neurodegenerative disorders
3.6. Amyloid-β and tau proteins deformities and microtubule organisation involving AD [134].
Microtubule stabilization is an essential measure of the normal
The most highlighted proteins in AD are tau and Aβ [127], their function of neurons. Defects in microtubule association have been
accumulation in AD brains is known to be a major hallmark [128]. Tau is studied in AD. A study by Brandt and Bacota states that adjusting the
a microtubule-associated cytosolic protein (MAP) found in the axonal microtubule dynamics can be considered a promising approach for AD
length of the neuron [129]. This protein plays a crucial role in stabilizing drugs [135]. On the other hand, Aβ peptide is known to form plaques
microtubules, maintaining DNA and overall neuronal health [130,131]. extracellularly [136]. These peptides at the required physiological
There are series of events that act as stimuli (chemical, physical or concentration ameliorate memory. Aβ aggregation induces neurotox­
infection) to initiate inflammation. Activation of microglial cells and icity that is dependent on NFTs formation. Therefore, the expression of
astrocytes deliver inflammatory mediators like nitric oxide (NO), TNF-α, Aβ plaques is not a distinguishing marker between natural aging and AD
PGs, IL-6 and ROS. These agents work collectively to initiate neuro­ brain [137,138]. However, few studies have supported the fact of
toxicity. One consequence is the disruption in ubiquitin-proteosome focusing tau and Aβ as potential target for drug therapy in AD [139,
cascade, which leads to the aggregation of proteins. These proteins, if 140]. Moreover, Aβ act as a stimulus for inflammation that excites glial
not cleared away then cells trigger neuroinflammation and apoptosis. cells to produce pro-inflammatory cytokines such as TNF-α, IL-6 and
However, proteolysis by caspase induces tau cleavage; this further leads IL-1β and inflammatory reaction protein like C-reactive protein (CRP).

Fig. 3. Schematic illustration of BA inhibiting/reducing the AChE activity in cholinergic pathway involved in AD pathogenesis. Ch- choline; ACh- acetylcholine; A-
acetate; a7nAchR- α7 nicotinic acetylcholine receptor; AChE- acetycholinesterase; TNF-α - tumor necrosis factor α; IL 6/1β- interleukin 6/1β; NF-kB- nuclear factor
kappa-light-chain-enhancer of activated B cells; Aβ- amyloid-beta; BA- boswellic acid.

6
A. Siddiqui et al. Biomedicine & Pharmacotherapy 144 (2021) 112250

These agents have the potential to further stimulate glial cells to release it was demonstrated that sublingually administered AKBA (5 mg/kg)
phosphorylated tau, Aβ42 and other pro-inflammatory mediators [141]. have reverted depressive like symptoms due to its anti-glutaminergic
On the other hand, enhanced IL-1β and other cytokines expression also and antioxidant properties. In addition, such treatment also caused a
trigger CDK5 to activate tau hyperphosphorylation which leads to the reduction in glial fibrillary acid protein (GFAP), glutamate (GLU),
formation of NFTs [27]. kynurenine (KYN) and NF-kB in hippocampus and prefrontal cortex
At present, most of the research studies have been focused on Aβ and region [46]. These results suggest further molecular investigations are
Tau pathology to achieve the goal of AD therapy [142–144]. A study required to explore the potential of BAs (Fig. 4) as a drug candidate
shows the effect of β-BA on the outgrowth of neurite and branching of against tau and Aβ associated pathophysiology.
hippocampus neurons by Karima et al. [145]. They reported that β-BA
has the potential to improve branching, the outgrowth of neurite, and 4. Future prospective
advances tubulin polymerization. Afterward, they studied the mecha­
nism of β-BA on the tubulin assembly and the results shows that βBA has In the last few decades, different pathological hallmarks involved in
significantly strengthened microtubule polymerization and increases the AD have been studied thoroughly. However, the target-oriented drug
length of microtubules, which protected microtubule disorientation and approach is still left out. The present review has discussed the patho­
axonal degradation as well. Therefore, it can be concluded that βBA physiological changes in the brain resulting from AD involving disrup­
could be a valuable agent against neurodegenerative diseases [146]. tion in Nrf2 and NF-kB crosstalk, ACh pathway, Aβ plaque accumulation
Another study in rodents demonstrated that AKBA downregulated the and, NFTs formation, ultimately leading to inflammation and cytotoxic
expression of BACE1, which is responsible for the cleavage of APP, mechanisms activation. Numerous compounds have shown their po­
therefore, inhibiting Aβ accumulation in AD brains. Simultaneously, tency alone or in combination against AD, such as donepezil, galant­
AKBA also exerts anti-inflammatory and antioxidant effects by inhibit­ amine, memantine, rivastigmine [151]. Natural compounds, on the
ing the release of inflammatory mediators [82]. other hand, are winning more attention as they have a therapeutic index
Many studies have been conducted so far, including the effect of with possibly no side effects and are affordable for long-run treatment in
terpenoids on Aβ plaques and neurofibrillary tangle formation [147, chronic diseases [152]. The plant derivatives have broadened the scope
148]. Similarly, another research study of α-BA on primary fetal human of the medicinal system. Likewise, a group of compounds known as
cell lines against streptozotocin-induced AD hallmarks in astrocytes such boswellic acids were considered as valuable as gold and used in religious
as hyperphosphorylated tau, cell death and ROS formation shows a ceremonies in olden days. Now, BAs have reattained its reputation due
significant potential decrease in tau hyperphosphorylation, ROS for­ to its pleiotropic biological activities. However, AKBA is one of the most
mation and improved cell proliferation [149]. A recent study demon­ potent bioactive constituent among BAs, which has shown assuring re­
strated anti-inflammatory effect of BAs in lipopolysaccharide (LPS) sults as a potential candidate against inflammation induced AD pathway
stimulated microglial cells via down regulating expression of inflam­ in basic research studies. Further investigations on boswellia extract’s
matory molecules such as IL-1β, IL-6, inducible nitric oxide synthase phytoconstituents including AKBA activity on different signaling path­
(iNOS) and NF-kB [150]. In the Aβ-induced rodent model of depression, ways are needed to be explored; also, there is a requirement to study its

Fig. 4. Represents the potential role of BA inhibiting Aβ plaques and NFT induced inflammatory and neurotoxicity mechanism in AD. NF-kB- nuclear factor kappa-
light-chain-enhancer of activated B cells; iNOS- inducible nitric oxide synthase; TNF-α - tumor necrosis factor a; IL-1β/6 - Interleukin 1β/6; ROS- reactive oxygen
species; BACE1- beta-site APP cleaving enzyme 1; APP- amyloid precursor protein; Aβ- amyloid-beta; CDK5- cyclin dependant kinase enzyme; P- phosphorylation;
BA- boswellic acid; AD- Alzheimer’s disease.

7
A. Siddiqui et al. Biomedicine & Pharmacotherapy 144 (2021) 112250

bioavailability, drug interactions and pharmacokinetic properties. Alzheimer’s disease: initial results of a co-twin control study, Neurology 44 (2)
(1994) 227–232.
[12] J.B. Rich, D.X. Rasmusson, M.F. Folstein, K.A. Carson, C. Kawas, J. Brandt,
5. Conclusion Nonsteroidal anti-inflammatory drugs in Alzheimer’s disease, Neurology 45 (1)
(1995) 51–55.
[13] P.L. McGeer, E.G. McGeer, NSAIDs and Alzheimer disease: epidemiological,
Based on evidences it is concluded that inflammation plays a crucial
animal model and clinical studies, Neurobiol. Aging 28 (5) (2007) 639–647.
role in AD pathophysiology. Research studies help in establishing that [14] M. Miguel-Álvarez, A. Santos-Lozano, F. Sanchis-Gomar, C. Fiuza-Luces,
inflammation is a response to protein accumulation in the brain and H. Pareja-Galeano, N. Garatachea, A. Lucia, Non-steroidal anti-inflammatory
drugs as a treatment for Alzheimer’s disease: a systematic review and meta-
vice-versa. After all these investigations, there are still limited treatment
analysis of treatment effect, Drugs Aging 32 (2) (2015) 139–147.
options available that have not been able to inhibit/modulate the [15] P.L. McGeer, J. Rogers, Anti-inflammatory agents as a therapeutic approach to
pathways involved in inflammation associated- neurodegeneration in Alzheimer’s disease, Neurology 42 (2) (1992) 447–449.
AD. On the other hand, boswellic acids are diverse in nature due to its [16] E. Zotova, J.A. Nicoll, R. Kalaria, C. Holmes, D. Boche, Inflammation in
Alzheimer’s disease: relevance to pathogenesis and therapy, Alzheimers Res Ther.
therapeutic efficacy in different types of diseases. In different AD 2 (1) (2010) 1.
models, boswellic acids have demonstrated beneficial effects via [17] A. Serrano-Pozo, M.L. Mielke, T. Gómez-Isla, R.A. Betensky, J.H. Growdon, M.
modulating the activity of multiple molecular targets and signaling P. Frosch, B.T. Hyman, Reactive glia not only associates with plaques but also
parallels tangles in Alzheimer’s disease, Am. J. Pathol. 179 (3) (2011)
pathways that contribute to the pathogenesis of AD. Further in­ 1373–1384.
vestigations of BAs in inflammation-induced neurodegeneration should [18] J.C. Lambert, C.A. Ibrahim-Verbaas, D. Harold, A.C. Naj, R. Sims, C. Bellenguez,
be explored to ascertain its pharmacological aspects. G. Jun, A.L. DeStefano, J.C. Bis, G.W. Beecham, B. Grenier-Boley, G. Russo, T.
A. Thornton-Wells, N. Jones, A.V. Smith, V. Chouraki, C. Thomas, M.A. Ikram,
D. Zelenika, B.N. Vardarajan, Y. Kamatani, C.F. Lin, A. Gerrish, H. Schmidt,
CRediT authorship contribution statement B. Kunkle, M.L. Dunstan, A. Ruiz, M.T. Bihoreau, S.H. Choi, C. Reitz, F. Pasquier,
P. Hollingworth, A. Ramirez, O. Hanon, A.L. Fitzpatrick, J.D. Buxbaum,
D. Campion, P.K. Crane, C. Baldwin, T. Becker, V. Gudnason, C. Cruchaga,
AS and YK carried out literature review, conceptualized, designed D. Craig, N. Amin, C. Berr, O.L. Lopez, P.L. De Jager, V. Deramecourt, J.
and drafted the manuscript. ZS, RNJ and IO have provided critical A. Johnston, D. Evans, S. Lovestone, L. Letenneur, F.J. Morón, D.C. Rubinsztein,
suggestions and corrected the final version of manuscript. All authors G. Eiriksdottir, K. Sleegers, A.M. Goate, N. Fiévet, M.J. Huentelman, M. Gill,
K. Brown, M.I. Kamboh, L. Keller, P. Barberger-Gateau, B. McGuinness, E.
read and approved the final manuscript. B. Larson, R. Green, A.J. Myers, C. Dufouil, S. Todd, D. Wallon, S. Love,
E. Rogaeva, J. Gallacher, P. St George-Hyslop, J. Clarimon, A. Lleo, A. Bayer, D.
W. Tsuang, L. Yu, M. Tsolaki, P. Bossù, G. Spalletta, P. Proitsi, J. Collinge,
Conflict of interest statement S. Sorbi, F. Sanchez-Garcia, N.C. Fox, J. Hardy, M.C.D. Naranjo, P. Bosco,
R. Clarke, C. Brayne, D. Galimberti, M. Mancuso, F. Matthews, S. Moebus,
The authors declare that they have no known competing financial P. Mecocci, M. Del Zompo, W. Maier, H. Hampel, A. Pilotto, M. Bullido, F. Panza,
P. Caffarra, B. Nacmias, J.R. Gilbert, M. Mayhaus, L. Lannfelt, H. Hakonarson,
interests or personal relationships that could have appeared to influence S. Pichler, M.M. Carrasquillo, M. Ingelsson, D. Beekly, V. Alvarez, F. Zou,
the work reported in this paper. O. Valladares, S.G. Younkin, E. Coto, K.L. Hamilton-Nelson, W. Gu, C. Razquin,
P. Pastor, I. Mateo, M.J. Owen, K.M. Faber, P.V. Jonsson, O. Combarros, M.
C. O’Donovan, L.B. Cantwell, H. Soininen, D. Blacker, S. Mead, T.H. Mosley, D.
Acknowledgements A. Bennett, T.B. Harris, L. Fratiglioni, C. Holmes, R.F.A.G. de Bruijn, P. Passmore,
T.J. Montine, K. Bettens, J.I. Rotter, A. Brice, K. Morgan, T.M. Foroud, W.
We thank Jeffrey Cheah School of Medicine and Health Sciences, A. Kukull, D. Hannequin, J.F. Powell, M.A. Nalls, Meta-analysis of 74,046
individuals identifies 11 new susceptibility loci for Alzheimer’s disease, Nat.
Monash University Malaysia for School Collaborative Grant 2021 (MED/ Genet. 45 (12) (2013) 1452–1458.
SCG2021/07; SED-000064), for the financial support to our work on the [19] X.L. Bu, X.Q. Yao, S.S. Jiao, F. Zeng, Y.H. Liu, Y. Xiang, C.R. Liang, Q.H. Wang,
neurodegeneration and possible alternative therapeutic targets, which X. Wang, H.Y. Cao, X. Yi, B. Deng, C.H. Liu, J. Xu, L.L. Zhang, C.Y. Gao, Z.Q. Xu,
M. Zhang, L. Wang, X.L. Tan, X. Xu, H.D. Zhou, Y.J. Wang, A study on the
was the starting point of this review article.
association between infectious burden and Alzheimer’s disease, Eur. J. Neurol. 22
(12) (2015) 1519–1525.
References [20] J. Budni, M. L. Garcez, J. de Medeiros, E. Cassaro, T. Bellettini-Santos, F. Mina,
J. Quevedo, The anti-inflammatory role of minocycline in Alzheimer´s disease,
Curr. Alzheimer Res. 13 (12) (2016) 1319–1329.
[1] International, A.s.D., World Alzheimer report 2019: attitudes to dementia.
[21] N. Zilka, Z. Kazmerova, S. Jadhav, P. Neradil, A. Madari, D. Obetkova, O. Bugos,
Alzheimer’s Disease International: London, 2019.
M. Novak, Who fans the flames of Alzheimer’s disease brains? Misfolded tau on
[2] AS Assication2019 Alzheimer’s disease facts and figures Alzheimer’s Dement.,
the crossroad of neurodegenerative and inflammatory pathways,
15, 3, 2019, pp. 321–387.
J. Neuroinflamm. 9 (2012) 47.
[3] K.E. Murphy, J.J. Park, Can co-activation of Nrf2 and neurotrophic signaling
[22] D. Tejera, M.T. Heneka, Microglia in Alzheimer’s disease: the good, the bad and
pathway slow Alzheimer’s disease? Int. J. Mol. Sci. 18 (6) (2017) 1168.
the ugly, Curr. Alzheimer Res. 13 (4) (2016) 370–380.
[4] M.W. Wong, N. Braidy, A. Poljak, R. Pickford, M. Thambisetty, P.S. Sachdev,
[23] B. Brugg, Y.L. Dubreuil, G. Huber, E.E. Wollman, N. Delhaye-Bouchaud,
Dysregulation of lipids in Alzheimer’s disease and their role as potential
J. Mariani, Inflammatory processes induce beta-amyloid precursor protein
biomarkers, Alzheimers Demen. 13 (7) (2017) 810–827.
changes in mouse brain, Proc. Natl. Acad. Sci. U.S.A. 92 (7) (1995) 3032–3035.
[5] G. Azizi, N. Khannazer, A. Mirshafiey, The potential role of chemokines in
[24] M. Fiala, D.H. Cribbs, M. Rosenthal, G. Bernard, Phagocytosis of amyloid-beta
Alzheimer’s disease pathogenesis, Am. J. Alzheimer Dis. Demen. 29 (5) (2014)
and inflammation: two faces of innate immunity in Alzheimer’s disease,
415–425.
J. Alzheimers Dis. 11 (4) (2007) 457–463.
[6] G. Azizi, A. Mirshafiey, The potential role of proinflammatory and
[25] D. Goldgaber, H.W. Harris, T. Hla, T. Maciag, R.J. Donnelly, J.S. Jacobsen, M.
antiinflammatory cytokines in Alzheimer disease pathogenesis,
P. Vitek, D.C. Gajdusek, Interleukin 1 regulates synthesis of amyloid beta-protein
Immunopharmacol. Immunotoxicol. 34 (6) (2012) 881–895.
precursor mRNA in human endothelial cells, Proc. Natl. Acad. Sci. U.S.A. 86 (19)
[7] W.S. Griffin, L.C. Stanley, C. Ling, L. White, V. MacLeod, L.J. Perrot, C.L. White,
(1989) 7606–7610.
C. Araoz, Brain interleukin 1 and S-100 immunoreactivity are elevated in Down
[26] B.L. Plassman, R.J. Havlik, D.C. Steffens, M.J. Helms, T.N. Newman, D. Drosdick,
syndrome and Alzheimer disease, Proc. Natl. Acad. Sci. U.S.A. 86 (19) (1989)
C. Phillips, B.A. Gau, K.A. Welsh-Bohmer, J.R. Burke, J.M. Guralnik, J.C.
7611–7615.
S. Breitner, Documented head injury in early adulthood and risk of Alzheimer’s
[8] J. Rogers, J. Luber-Narod, S.D. Styren, W.H. Civin, Expression of immune system-
disease and other dementias, Neurology 55 (8) (2000) 1158–1166.
associated antigens by cells of the human central nervous system: relationship to
[27] R.A. Quintanilla, D.I. Orellana, C. González-Billault, R.B. Maccioni, Interleukin-6
the pathology of Alzheimer’s disease, Neurobiol. Aging 9 (1988) 339–349.
induces Alzheimer-type phosphorylation of tau protein by deregulating the cdk5/
[9] T. Wyss-Coray, F. Yan, A.H. Lin, J.D. Lambris, J.J. Alexander, R.J. Quigg,
p35 pathway, Exp. Cell Res 295 (1) (2004) 245–257.
E. Masliah, Prominent neurodegeneration and increased plaque formation in
[28] T. Wyss-Coray, J. Rogers, Inflammation in Alzheimer disease—A brief review of
complement-inhibited Alzheimer’s mice, Proc. Natl. Acad. Sci. U.S.A. 99 (16)
the basic science and clinical literature, Cold Spring Harb. Perspect. Med. 2 (1)
(2002) 10837–10842.
(2012), a006346.
[10] C.M. Beard, S.C. Waring, P.C. O’Brien, L.T. Kurland, E. Kokmen, Nonsteroidal
[29] A. Qaseem, V. Snow, Jr Cross JT, M.A. Forciea, Jr Hopkins R, P. Shekelle,
anti-inflammatory drug use and Alzheimer’s disease: a case-control study in
A. Adelman, D. Mehr, K. Schellhase, D. Campos-Outcalt, P. Santaguida, D.
Rochester, Minnesota, 1980 through 1984, Mayo Clin. Proc. 73 (10) (1998)
K. Owens, D. American College of Physicians/American Academy of Family
951–955.
Physicians Panel on, Current pharmacologic treatment of dementia: a clinical
[11] J.C. Breitner, B.A. Gau, K.A. Welsh, B.L. Plassman, W.M. McDonald, M.J. Helms,
J.C. Anthony, Inverse association of anti-inflammatory treatments and

8
A. Siddiqui et al. Biomedicine & Pharmacotherapy 144 (2021) 112250

practice guideline from the American College of Physicians and the American [56] S. Chakraborty, M. Lakshmanan, H.L.F. Swa, J. Chen, X. Zhang, Y.S. Ong, L.
Academy of Family Physicians, Ann. Intern. Med. 148 (5) (2008) 370–378. S. Loo, S.C. Akıncılar, J. Gunaratne, V. Tergaonkar, K.M. Hui, W. Hong, An
[30] D. Jaturapatporn, M.G.E.K.N. Isaac, J. McCleery, N. Tabet, Aspirin, steroidal and oncogenic role of Agrin in regulating focal adhesion integrity in hepatocellular
non-steroidal anti-inflammatory drugs for the treatment of Alzheimer’s disease, carcinoma, Nat. Commun. 6 (1) (2015) 1–16.
Cochrane Database Syst. Rev. 2 (2012), Cd006378. [57] Y. Li, H.S. Cheng, W.J. Chng, V. Tergaonkar, Activation of mutant TERT promoter
[31] B. Dunn, P. Stein, P. Cavazzoni, Approval of aducanumab for Alzheimer by RAS-ERK signaling is a key step in malignant progression of BRAF-mutant
disease—The FDA’s perspective, JAMA Intern. Med. (2021). human melanomas, Proc. Natl. Acad. Sci. U.S.A. 113 (50) (2016) 14402–14407.
[32] J. Sevigny, P. Chiao, T. Bussière, P.H. Weinreb, L. Williams, M. Maier, R. Dunstan, [58] D. Poeckel, O. Werz, Boswellic acids: biological actions and molecular targets,
S. Salloway, T. Chen, Y. Ling, J. O’Gorman, F. Qian, M. Arastu, M. Li, S. Chollate, Curr. Med. Chem. 13 (28) (2006) 3359–3369.
M.S. Brennan, O. Quintero-Monzon, R.H. Scannevin, H.M. Arnold, T. Engber, [59] H. Safayhi, et al., Inhibition by boswellic acids of human leukocyte elastase,
K. Rhodes, J. Ferrero, Y. Hang, A. Mikulskis, J. Grimm, C. Hock, R.M. Nitsch, J. Pharmacol. Exp. Ther. 281 (1) (1997) 460–463.
A. Sandrock, The antibody aducanumab reduces Aβ plaques in Alzheimer’s [60] E.R. Sailer, L.R. Subramanian, B. Rall, R.F. Hoernlein, H.P.T. Ammon, H. Safayhi,
disease, Nature 537 (7618) (2016) 50–56. Acetyl-11–keto-β-boswellic acid (AKBA): structure requirements for binding and
[33] S. Moghadamtousi, B. Goh, C. Chan, T. Shabab, H. Kadir, Biological activities and 5–lipoxygenase inhibitory activity, Br. J. Pharmacol. 117 (4) (1996) 615–618.
phytochemicals of Swietenia macrophylla King, Molecules 18 (9) (2013) [61] H. Ammon, Modulation of the immune system by Boswellia serrata extracts and
10465–10483. boswellic acids, Phytomedicine 17 (11) (2010) 862–867.
[34] Y.S. Park, J.H. Lee, J. Bondar, J.A. Harwalkar, H. Safayhi, M. Golubic, Cytotoxic [62] M. Bishnoi, C.S. Patil, A. Kumar, S.K. Kulkarni, Protective effects of nimesulide
action of acetyl-11-keto-beta-boswellic acid (AKBA) on meningioma cells, Planta (COX inhibitor), AKBA (5-LOX inhibitor), and their combination in aging-
Med. 68 (5) (2002) 397–401. associated abnormalities in mice, Methods Find. Exp. Clin. Pharmacol. 27 (7)
[35] M.Z. Siddiqui, Boswellia serrata, a potential antiinflammatory agent: an (2005) 465–470.
overview, Indian J. Pharm. Sci. 73 (3) (2011) 255–261. [63] O. Werz, 5-lipoxygenase: cellular biology and molecular pharmacology, Curr.
[36] B. Büchele, W. Zugmaier, T. Simmet, Analysis of pentacyclic triterpenic acids Drug Targets-Inflamm. Allergy 1 (1) (2002) 23–44.
from frankincense gum resins and related phytopharmaceuticals by high- [64] A. Di Gennaro, C. Carnini, C. Buccellati, R. Ballerio, S. Zarini, F. Fumagalli,
performance liquid chromatography. Identification of lupeolic acid, a novel S. Viappiani, L. Librizzi, A. Hernandez, R.C. Murphy, G. Constantin, M. De Curtis,
pentacyclic triterpene, J. Chromatogr. B Anal. Technol. Biomed. Life Sci. 791 G. Folco, A. Sala, Cysteinyl-leukotriene receptor activation in brain inflammatory
(1–2) (2003) 21–30. reactions and cerebral edema formation: a role for transcellular biosynthesis of
[37] H. Ammon, Boswellic acids and their role in chronic inflammatory diseases, Anti- cysteinyl leukotrienes, FASEB J. 18 (7) (2004) 842–844.
Inflamm. Nutraceuticals Chronic Dis. (2016) 291–327. [65] J. Michael, J. Marschallinger, L. Aigner, The leukotriene signaling pathway: a
[38] B. Büchele, T. Simmet, Analysis of 12 different pentacyclic triterpenic acids from druggable target in Alzheimer’s disease, Drug Discov. Today 24 (2) (2019)
frankincense in human plasma by high-performance liquid chromatography and 505–516.
photodiode array detection, J. Chromatogr. B 795 (2) (2003) 355–362. [66] O. Firuzi, J. Zhuo, C.M. Chinnici, T. Wisniewski, D. Praticò, 5–Lipoxygenase gene
[39] F. Iram, S.A. Khan, A. Husain, Phytochemistry and potential therapeutic actions disruption reduces amyloid-β pathology in a mouse model of Alzheimer’s disease,
of Boswellic acids: a mini-review, Asian Pac. J. Trop. Biomed. 7 (6) (2017) FASEB J. 22 (4) (2008) 1169–1178.
513–523. [67] M.D. Ikonomovic, E.E. Abrahamson, T. Uz, H. Manev, S.T. DeKosky, Increased 5-
[40] H. Ammon, Boswellic acids in chronic inflammatory diseases, Planta Med. 72 (12) lipoxygenase immunoreactivity in the hippocampus of patients with Alzheimer’s
(2006) 1100–1116. disease, J. Histochem. Cytochem. 56 (12) (2008) 1065–1073.
[41] M. Takahashi, B. Sung, Y. Shen, K. Hur, A. Link, C.R. Boland, B.B. Aggarwal, [68] M. Verhoff, S. Seitz, M. Paul, S.M. Noha, J. Jauch, D. Schuster, O. Werz, Tetra-and
A. Goel, Boswellic acid exerts antitumor effects in colorectal cancer cells by pentacyclic triterpene acids from the ancient anti-inflammatory remedy
modulating expression of the let-7 and miR-200 microRNA family, Carcinogenesis frankincense as inhibitors of microsomal prostaglandin E2 synthase-1, J. Nat.
33 (12) (2012) 2441–2449. Prod. 77 (6) (2014) 1445–1451.
[42] R. Hamidpour, S. Hamidpour, M. Hamidpour, M. Shahlari, Frankincense (乳香 Rǔ [69] U. Siemoneit, B. Hofmann, N. Kather, T. Lamkemeyer, J. Madlung, L. Franke,
Xiāng; Boswellia species): from the selection of traditional applications to the G. Schneider, J. Jauch, D. Poeckel, O. Werz, Identification and functional analysis
novel phytotherapy for the prevention and treatment of serious diseases. of cyclooxygenase-1 as a molecular target of boswellic acids, Biochem.
J. Tradit. Complement. Med. 3 (4) (2013) 221–226. Pharmacol. 75 (2) (2008) 503–513.
[43] Z. Du, Z. Liu, Z. Ning, Y. Liu, Z. Song, C. Wang, A. Lu, Prospects of boswellic acids [70] U. Siemoneit, A. Koeberle, A. Rossi, F. Dehm, M. Verhoff, S. Reckel, T. Maier,
as potential pharmaceutics, Planta Med. 81 (04) (2015) 259–271. J. Jauch, H. Northoff, F. Bernhard, V. Doetsch, L. Sautebin, O. Werz, Inhibition of
[44] K. Gerbeth, J. Hüsch, G. Fricker, O. Werz, M. Schubert-Zsilavecz, M. Abdel- microsomal prostaglandin E2 synthase-1 as a molecular basis for the anti-
Tawab, In vitro metabolism, permeation, and brain availability of six major inflammatory actions of boswellic acids from frankincense, Br. J. Pharmacol. 162
boswellic acids from Boswellia serrata gum resins, Fitoterapia 84 (2013) 99–106. (1) (2011) 147–162.
[45] K. Reising, J. Meins, B. Bastian, G. Eckert, W.E. Mueller, M. Schubert-Zsilavecz, [71] A.S. Sayed, N.S.E.D. El Sayed, Co-administration of 3-acetyl-11-keto-beta-bos­
M. Abdel-Tawab, Determination of boswellic acids in brain and plasma by high- wellic acid potentiates the protective effect of celecoxib in lipopolysaccharide-
performance liquid chromatography/tandem mass spectrometry, Anal. Chem. 77 induced cognitive impairment in mice: possible implication of anti-inflammatory
(20) (2005) 6640–6645. and antiglutamatergic pathways, J. Mol. Neurosci. 59 (1) (2016) 58–67.
[46] M.G. Morgese, M. Bove, M. Francavilla, S. Schiavone, S. Dimonte, A.L. Colia, [72] N. Marefati, F. Beheshti, S. Memarpour, R. Bayat, M. Naser Shafei, H.
M. Bevilacqua, L. Trabace, P. Tucci, Sublingual AKBA exerts antidepressant R. Sadeghnia, H. Ghazavi, M. Hosseini, The effects of acetyl-11-keto-β-boswellic
effects in the Aβ-treated mouse model, Biomolecules 11 (5) (2021) 686. acid on brain cytokines and memory impairment induced by lipopolysaccharide
[47] S. Sharma, V. Thawani, L. Hingorani, M. Shrivastava, V.R. Bhate, R. Khiyani, in rats, Cytokine 131 (2020), 155107.
Pharmacokinetic study of 11-keto β-Boswellic acid, Phytomedicine 11 (2–3) [73] S. Choi, J. Krishnan, K. Ruckmani, Cigarette smoke and related risk factors in
(2004) 255–260. neurological disorders: an update, Biomed. Pharmacother. 85 (2017) 79–86.
[48] A.R. Al-Yasiry, B. Kiczorowska, Frankincense–therapeutic properties, Post. Hig. [74] E.E. Vomhof-DeKrey, M.J. Picklo Sr., The Nrf2-antioxidant response element
Med. Dosw 70 (2016) 380–391. pathway: a target for regulating energy metabolism, J. Nutr. Biochem. 23 (10)
[49] M. Takahashi, B. Sung, Y. Shen, K. Hur, A. Link, C.R. Boland, B.B. Aggarwal, (2012) 1201–1206.
A. Goel, Boswellic acid exerts antitumor effects in colorectal cancer cells by [75] A. Cuadrado, G. Manda, A. Hassan, M.J. Alcaraz, C. Barbas, A. Daiber, P. Ghezzi,
modulating expression of the let-7 and miR-200 microRNA family, Carcinogenesis R. León, M.G. López, B. Oliva, M. Pajares, A.I. Rojo, N. Robledinos-Antón, A.
33 (12) (2012) 2441–2449. M. Valverde, E. Guney, H. Schmidt, Transcription factor NRF2 as a therapeutic
[50] R. Hamidpour, S. Hamidpour, M. Hamidpour, M. Shahlari, Frankincense ( rǔ target for chronic diseases: a systems medicine approach, Pharmacol. Rev. 70 (2)
xiāng; boswellia species): from the selection of traditional applications to the (2018) 348–383.
novel phytotherapy for the prevention and treatment of serious diseases, [76] M. Sandberg, J. Patil, B. D’Angelo, S.G. Weber, C. Mallard, NRF2-regulation in
J. Tradit. Complement. Med. 3 (4) (2013) 221–226. brain health and disease: implication of cerebral inflammation,
[51] D. Bordoloi, K. Banik, B. Shabnam, G. Padmavathi, J. Monisha, F. Arfuso, Neuropharmacology 79 (2014) 298–306.
A. Dharmarajan, X. Mao, L. Lim, L. Wang, L. Fan, K. Hui, A. Kumar, G. Sethi, [77] R.K. Sajja, S. Prasad, S. Tang, M.A. Kaisar, L. Cucullo, Blood-brain barrier
A. Kunnumakkara, TIPE family of proteins and its implications in different disruption in diabetic mice is linked to Nrf2 signaling deficits: role of ABCB10?
chronic diseases, Int. J. Mol. Sci. 19 (10) (2018) 2974. Neurosci. Lett. 653 (2017) 152–158.
[52] A.B. Kunnumakkara, D. Bordoloi, G. Padmavathi, J. Monisha, N.K. Roy, S. Prasad, [78] R.A. Kowluru, M. Mishra, Epigenetic regulation of redox signaling in diabetic
B.B. Aggarwal, Curcumin, the golden nutraceutical: multitargeting for multiple retinopathy: role of Nrf2, Free Radic. Biol. Med. 103 (2017) 155–164.
chronic diseases, Br. J. Pharmacol. 174 (11) (2017) 1325–1348. [79] A.T. Dinkova-Kostova, A.Y. Abramov, The emerging role of Nrf2 in mitochondrial
[53] N.K. Roy, A. Deka, D. Bordoloi, S. Mishra, A.P. Kumar, G. Sethi, A. function, Free Radic. Biol. Med. 88 (2015) 179–188.
B. Kunnumakkara, The potential role of boswellic acids in cancer prevention and [80] R.K. Sajja, K.N. Green, L. Cucullo, Altered nrf2 signaling mediates hypoglycemia-
treatment, Cancer Lett. 377 (1) (2016) 74–86. induced blood–brain barrier endothelial dysfunction in vitro, PLoS One 10 (3)
[54] D. Wang, et al., Boswellic acid exerts potent anticancer effects in HCT-116 human (2015), 0122358.
colon cancer cells mediated via induction of apoptosis, cell cycle arrest, cell [81] A. Silva-Palacios, M. Ostolga-Chavarría, C. Zazueta, M. Königsberg, Nrf2:
migration inhibition and inhibition of PI3K/AKT signalling pathway, J. BU .: Off. molecular and epigenetic regulation during aging, Ageing Res. Rev. 47 (2018)
J. Balk. Union Oncol. 23 (2) (2018) 340–345. 31–40.
[55] S.C. Akıncılar, K.C. Low, C.Y. Liu, T.D. Yan, A. Oji, M. Ikawa, S. Li, V. Tergaonkar, [82] C. Wei, J. Fan, X. Sun, J. Yao, Y. Guo, B. Zhou, Y. Shang, Acetyl-11-keto-
Quantitative assessment of telomerase components in cancer cell lines, FEBS Lett. β-boswellic acid ameliorates cognitive deficits and reduces amyloid-β levels in
589 (9) (2015) 974–984.

9
A. Siddiqui et al. Biomedicine & Pharmacotherapy 144 (2021) 112250

APPswe/PS1dE9 mice through antioxidant and anti-inflammatory pathways, Free [110] E. Nizri, Y. Hamra-Amitay, C. Sicsic, I. Lavon, T. Brenner, Anti-inflammatory
Radic. Biol. Med. 150 (2020) 96–108. properties of cholinergic up-regulation: a new role for acetylcholinesterase
[83] K.M. Holmström, L. Baird, Y. Zhang, I. Hargreaves, A. Chalasani, J.M. Land, inhibitors, Neuropharmacology 50 (5) (2006) 540–547.
L. Stanyer, M. Yamamoto, A.T. Dinkova-Kostova, A.Y. Abramov, Nrf2 impacts [111] F. Burčul, I. Generalić Mekinić, M. Radan, P. Rollin, I. Blažević, Isothiocyanates:
cellular bioenergetics by controlling substrate availability for mitochondrial cholinesterase inhibiting, antioxidant, and anti-inflammatory activity, J. Enzym.
respiration, Biol. Open 2 (8) (2013) 761–770. Inhib. Med. Chem. 33 (1) (2018) 577–582.
[84] T. Li, H. Wang, Y. Ding, M. Zhou, X. Zhou, X. Zhang, K. Ding, J. He, X. Lu, J. Xu, [112] M. Kamal, N. Greig, M. Reale, Anti-inflammatory properties of
W. Wei, Genetic elimination of Nrf2 aggravates secondary complications except acetylcholinesterase inhibitors administred in Alzheimer’s disease, Anti-Inflamm.
for vasospasm after experimental subarachnoid hemorrhage in mice, Brain Res. Anti-Allergy Agents Med. Chem. 8 (1) (2009) 85–100.
1558 (2014) 90–99. [113] N. Tabet, Acetylcholinesterase inhibitors for Alzheimer’s disease: anti-
[85] Y. Mitsuishi, K. Taguchi, Y. Kawatani, T. Shibata, T. Nukiwa, H. Aburatani, inflammatories in acetylcholine clothing!, Age Ageing 35 (4) (2006) 336–338.
M. Yamamoto, H. Motohashi, Nrf2 redirects glucose and glutamine into anabolic [114] S. Beheshti, R. Aghaie, Therapeutic effect of frankincense in a rat model of
pathways in metabolic reprogramming, Cancer Cell 22 (1) (2012) 66–79. Alzheimer’s disease, Avicenna J. Phytomed. 6 (4) (2016) 468–475.
[86] J.-M. Lee, A.Y. Shih, T.H. Murphy, J.A. Johnson, NF-E2-related factor-2 mediates [115] N.C. Inestrosa, A. Alvarez, J. Godoy, A. Reyes, G.V. De Ferrari,
neuroprotection against mitochondrial complex I inhibitors and increased Acetylcholinesterase–amyloid-β-peptide interaction and Wnt signaling
concentrations of intracellular calcium in primary cortical neurons, J. Biol. Chem. involvement in Aβ neurotoxicity, Acta Neurol. Scand. 102 (2000) 53–59.
278 (39) (2003) 37948–37956. [116] E. Viayna, R. Sabate, D. Muñoz-Torrero, Dual inhibitors of β-amyloid aggregation
[87] N.G. Innamorato, A.I. Rojo, Á.J. García-Yagüe, M. Yamamoto, M.L. de Ceballos, and acetylcholinesterase as multi-target anti-Alzheimer drug candidates, Curr.
A. Cuadrado, The transcription factor Nrf2 is a therapeutic target against brain Top. Med. Chem. 13 (15) (2013) 1820–1842.
inflammation, J. Immunol. 181 (1) (2008) 680–689. [117] P. Williams, A. Sorribas, M.-J.R. Howes, Natural products as a source of
[88] J. Tan, Q.X. Li, G. Evin, Effects of mild and severe oxidative stress on BACE1 Alzheimer’s drug leads, Nat. Prod. Rep. 28 (1) (2011) 48–77.
expression and APP amyloidogenic processing. Systems Biology of Alzheimer’s [118] V.A. Pavlov, K.J. Tracey, The cholinergic anti-inflammatory pathway, Brain
Disease, Springer, 2016, pp. 101–116. Behav. Immun. 19 (6) (2005) 493–499.
[89] P.L. McGeer, J. Rogers, E.G. McGeer, Inflammation, antiinflammatory agents, and [119] H. Wang, H. Liao, M. Ochani, M. Justiniani, X. Lin, L. Yang, Y. Al-Abed, H. Wang,
Alzheimer’s disease: the last 22 years, J. Alzheimers Dis. 54 (3) (2016) 853–857. C. Metz, E.J. Miller, K.J. Tracey, L. Ulloa, Cholinergic agonists inhibit HMGB1
[90] G. Joshi, K.A. Gan, D.A. Johnson, J.A. Johnson, Increased Alzheimer’s release and improve survival in experimental sepsis, Nat. Med. 10 (11) (2004)
disease–like pathology in the APP/PS1ΔE9 mouse model lacking Nrf2 through 1216–1221.
modulation of autophagy, Neurobiol. Aging 36 (2) (2015) 664–679. [120] Q. Li, X.D. Zhou, V.P. Kolosov, J.M. Perelman, Nicotine reduces TNF-α expression
[91] C. Branca, E. Ferreira, T.V. Nguyen, K. Doyle, A. Caccamo, S. Oddo, Genetic through a α7 nAChR/MyD88/NF-ĸB pathway in HBE16 airway epithelial cells,
reduction of Nrf2 exacerbates cognitive deficits in a mouse model of Alzheimer’s Cell. Physiol. Biochem. 27 (5) (2011) 605–612.
disease, Hum. Mol. Genet. 26 (24) (2017) 4823–4835. [121] S. Darvesh, R. Walsh, R. Kumar, A. Caines, S. Roberts, D. Magee, K. Rockwood,
[92] I. Buendia, P. Michalska, E. Navarro, I. Gameiro, J. Egea, R. León, Nrf2–ARE E. Martin, Inhibition of human cholinesterases by drugs used to treat Alzheimer
pathway: an emerging target against oxidative stress and neuroinflammation in disease, Alzheimer Dis. Assoc. Disord. 17 (2) (2003) 117–126.
neurodegenerative diseases, Pharmacol. Ther. 157 (2016) 84–104. [122] A.N. Syad, K.P. Devi, Botanics: a potential source of new therapies for Alzheimer’s
[93] M.P. Mattson, S. Camandola, NF-κB in neuronal plasticity and neurodegenerative disease? Bot.: Targets Ther. 4 (2014) 11–26.
disorders, J. Clin. Investig. 107 (3) (2001) 247–254. [123] T.M. Mohamed, M. Youssef, A.A. Bakry, M.M. El-Keiy, Alzheimer’s disease
[94] A. Oeckinghaus, S. Ghosh, The NF-kappaB family of transcription factors and its improved through the activity of mitochondrial chain complexes and their gene
regulation, Cold Spring Harb. Perspect. Biol. 1 (4) (2009), 000034. expression in rats by boswellic acid, Metab. Brain Dis. 36 (2) (2021) 255–264.
[95] J.S. Tilstra, A.R. Robinson, J. Wang, S.Q. Gregg, C.L. Clauson, D.P. Reay, L. [124] S. Ebrahimpour, M. Fazeli, S. Mehri, M. Taherianfard, H. Hosseinzadeh, Boswellic
A. Nasto, C.M. St Croix, A. Usas, N. Vo, J. Huard, P.R. Clemens, D.B. Stolz, D. acid improves cognitive function in a rat model through its antioxidant activity:-
C. Guttridge, S.C. Watkins, G.A. Garinis, Y. Wang, L.J. Niedernhofer, P. neuroprotective effect of boswellic acid, J. Pharmacopunct. 20 (1) (2017) 10–17.
D. Robbins, NF-κB inhibition delays DNA damage–induced senescence and aging [125] B. Alessandri, R.E. FitzGerald, U. Schaeppi, G.J. Krinke, W. Classen, The use of an
in mice, J. Clin. Investig. 122 (7) (2012) 2601–2612. unbaited tunnel maze in neurotoxicology: I. Trimethyltin-induced brain lesions,
[96] Q. Zhang, M.J. Lenardo, D. Baltimore, 30 years of NF-κB: a blossoming of Neurotoxicology 15 (2) (1994) 349–357.
relevance to human pathobiology, Cell 168 (1–2) (2017) 37–57. [126] H. Swartzwelder, Imparied maze performance in the rat caused by trimethyltin
[97] T. Hla, A. Ristimäki, S. Appleby, J.G. Barriocanal, Cyclooxygenase gene treatment: problem-solving deficits and perseveration, Neurobehav. Toxicol.
expression in inflammation and Angiogenesis a, Ann. N.Y. Acad. Sci. 696 (1) Teratol. 4 (1982) 169–176.
(1993) 197–204. [127] Y. Huang, L. Mucke, Alzheimer mechanisms and therapeutic strategies, Cell 148
[98] K. Sambamurti, R. Kinsey, B. Maloney, Y.W. Ge, D.K. Lahiri, Gene structure and (6) (2012) 1204–1222.
organization of the human β-secretase (BACE) promoter, FASEB J. 18 (9) (2004) [128] A. Boutajangout, E.M. Sigurdsson, P.K. Krishnamurthy, Tau as a therapeutic
1034–1036. target for Alzheimer’s disease, Curr. Alzheimer Res. 8 (6) (2011) 666–677.
[99] Y.J. Lee, I.S. Choi, M.H. Park, Y.M. Lee, J.K. Song, Y.H. Kim, K.H. Kim, D. [129] R.L. Neve, P. Harris, K.S. Kosik, D.M. Kurnit, T.A. Donlon, Identification of cDNA
Y. Hwang, J.H. Jeong, Y.P. Yun, K.W. Oh, J.K. Jung, S.B. Han, J.T. Hong, 4-O- clones for the human microtubule-associated protein tau and chromosomal
Methylhonokiol attenuates memory impairment in presenilin 2 mutant mice localization of the genes for tau and microtubule-associated protein 2, Mol. Brain
through reduction of oxidative damage and inactivation of astrocytes and the ERK Res. 1 (3) (1986) 271–280.
pathway, Free Radic. Biol. Med. 50 (1) (2011) 66–77. [130] J. Avila, J.S. Jiménez, C.L. Sayas, M. Bolós, J.C. Zabala, G. Rivas, F. Hernández,
[100] J.D. Wardyn, A.H. Ponsford, C.M. Sanderson, Dissecting molecular cross-talk Tau structures, Front. Aging Neurosci. 8 (2016) 262.
between Nrf2 and NF-κB response pathways, Biochem. Soc. Trans. 43 (4) (2015) [131] Y. Wang, E. Mandelkow, Tau in physiology and pathology, Nat. Rev. Neurosci. 17
621–626. (1) (2016) 5–21.
[101] V. Ganesh Yerra, G. Negi, S.S. Sharma, A. Kumar, Potential therapeutic effects of [132] M.J. Metcalfe, M.E. Figueiredo-Pereira, Relationship between tau pathology and
the simultaneous targeting of the Nrf2 and NF-κB pathways in diabetic neuroinflammation in Alzheimer’s disease, Mt. Sinai J. Med.: A J. Transl. Pers.
neuropathy, Redox Biol. 1 (1) (2013) 394–397. Med.: A J. Transl. Pers. Med. 77 (1) (2010) 50–58.
[102] A. Daverey, S.K. Agrawal, Curcumin protects against white matter injury through [133] E.M. Mandelkow, E. Mandelkow, Biochemistry and cell biology of tau protein in
NF-κB and Nrf2 cross talk, J. Neurotrauma 37 (10) (2020) 1255–1265. neurofibrillary degeneration, Cold Spring Harb. Perspect. Med. 2 (7) (2012),
[103] M.P. Soares, M.P. Seldon, I.P. Gregoire, T. Vassilevskaia, P.O. Berberat, J. Yu, T. 006247.
Y. Tsui, F.H. Bach, Heme oxygenase-1 modulates the expression of adhesion [134] L.C. Kapitein, C.C. Hoogenraad, Building the neuronal microtubule cytoskeleton,
molecules associated with endothelial cell activation, J. Immunol. 172 (6) (2004) Neuron 87 (3) (2015) 492–506.
3553–3563. [135] R. Brandt, L. Bakota, Microtubule dynamics and the neurodegenerative triad of
[104] G.-H. Liu, J. Qu, X. Shen, NF-κB/p65 antagonizes Nrf2-ARE pathway by depriving Alzheimer’s disease: the hidden connection, J. Neurochem. 143 (4) (2017)
CBP from Nrf2 and facilitating recruitment of HDAC3 to MafK, Biochim. Et 409–417.
Biophys. Acta (BBA)-Mol. Cell Res. 1783 (5) (2008) 713–727. [136] G. Ramachandran, J.B. Udgaonkar, Mechanistic studies unravel the complexity
[105] S.-W. Kim, H.K. Lee, J.H. Shin, J.K. Lee, Up-down regulation of HO-1 and iNOS inherent in tau aggregation leading to Alzheimer’s disease and the tauopathies,
gene expressions by ethyl pyruvate via recruiting p300 to Nrf2 and depriving It Biochemistry 52 (24) (2013) 4107–4126.
from p65, Free Radic. Biol. Med. 65 (2013) 468–476. [137] G.A. Czapski, K. Czubowicz, J.B. Strosznajder, R.P. Strosznajder, The
[106] Y. Ding, et al., Neuroprotection by acetyl-11-keto-β-boswellic acid, in ischemic lipoxygenases: their regulation and implication in Alzheimer’s disease,
brain injury involves the Nrf2/HO-1 defense pathway, Sci. Rep. 4 (1) (2014) 1–9. Neurochem. Res. 41 (1–2) (2016) 243–257.
[107] Y. Ding, M. Chen, M. Wang, Y. Li, A. Wen, Posttreatment with 11-keto-β-boswellic [138] Y. Akitake, Y. Nakatani, D. Kamei, M. Hosokawa, H. Akatsu, S. Uematsu, S. Akira,
acid ameliorates cerebral ischemia–reperfusion injury: Nrf2/HO-1 pathway as a I. Kudo, S. Hara, M. Takahashi, Microsomal prostaglandin E synthase-1 is induced
potential mechanism, Mol. Neurobiol. 52 (3) (2015) 1430–1439. in alzheimer’s disease and its deletion mitigates alzheimer’s disease-like
[108] T. Babic, The cholinergic hypothesis of Alzheimer’s disease: a review of progress, pathology in a mouse model, J. Neurosci. Res. 91 (7) (2013) 909–919.
J. Neurol., Neurosurg. Psychiatry 67 (4) (1999), 558-558. [139] J. Laurén, D.A. Gimbel, H.B. Nygaard, J.W. Gilbert, S.M. Strittmatter, Cellular
[109] Y. Yoshiyama, A. Kojima, C. Ishikawa, K. Arai, Anti-inflammatory action of prion protein mediates impairment of synaptic plasticity by amyloid-β oligomers,
donepezil ameliorates tau pathology, synaptic loss, and neurodegeneration in a Nature 457 (7233) (2009) 1128–1132.
tauopathy mouse model, J. Alzheimers Dis. 22 (1) (2010) 295–306. [140] E. Giacobini, G. Gold, Alzheimer disease therapy—moving from amyloid-β to tau,
Nat. Rev. Neurol. 9 (12) (2013) 677–686.

10
A. Siddiqui et al. Biomedicine & Pharmacotherapy 144 (2021) 112250

[141] S.M. Chiroma, M.T.H. Baharuldin, C.N.M. Taib, Z. Amom, S. Jagadeesan, M.A. [148] A.A. Gomaa, R.M. Makboul, M.A. Al-Mokhtar, M.A. Nicola, Polyphenol-rich
M. Moklas, Inflammation in Alzheimer’s disease: a friend or foe? Biomed. Res. Boswellia serrata gum prevents cognitive impairment and insulin resistance of
Ther. 5 (8) (2018) 2552–2564. diabetic rats through inhibition of GSK3β activity, oxidative stress and pro-
[142] T. Hamaguchi, K. Ono, M. Yamada, REVIEW: curcumin and Alzheimer’s disease, inflammatory cytokines, Biomed. Pharmacother. 109 (2019) 281–292.
CNS Neurosci. Ther. 16 (5) (2010) 285–297. [149] E. Fathi, F.H. Katouli, G.H. Riazi, M.D. Shasaltaneh, E. Parandavar, S. Bayati,
[143] H.J. Wobst, A. Sharma, M.I. Diamond, E.E. Wanker, J. Bieschke, The green tea A. Afrasiabi, R. Nazari, The effects of alpha boswellic acid on reelin expression
polyphenol (-)-epigallocatechin gallate prevents the aggregation of tau protein and tau phosphorylation in human astrocytes, Neuromol. Med. 19 (1) (2017)
into toxic oligomers at substoichiometric ratios, FEBS Lett. 589 (1) (2015) 77–83. 136–146.
[144] D.W. Peterson, R.C. George, F. Scaramozzino, N.E. LaPointe, R.A. Anderson, D. [150] X.-d Miao, L.J. Zheng, Z.Z. Zhao, S.L. Su, Y. Zhu, J.M. Guo, E.X. Shang, D.W. Qian,
J. Graves, J. Lew, Cinnamon extract inhibits tau aggregation associated with J.A. Duan, Protective effect and mechanism of boswellic acid and myrrha
Alzheimer’s disease in vitro, J. Alzheimers Dis. 17 (3) (2009) 585–597. sesquiterpenes with different proportions of compatibility on neuroinflammation
[145] O. Karima, G. Riazi, R. Yousefi, A.A. Movahedi, The enhancement effect of beta- by LPS-induced BV2 cells combined with network pharmacology, Molecules 24
boswellic acid on hippocampal neurites outgrowth and branching (an in vitro (21) (2019) 3946.
study), Neurol. Sci. 31 (3) (2010) 315–320. [151] D.A. Casey, D. Antimisiaris, J. O’Brien, Drugs for Alzheimer’s disease: are they
[146] O. Karima, G. Riazi, S. Khodadadi, R. Yousefi, K. Mahnam, F. Mokhtari, effective? Pharm. Ther. 35 (4) (2010) 208–211.
T. Cheraghi, E. Hoveizi, A.A. Moosavi-Movahedi, An in vitro study of the role of [152] A. Deorukhkar, S. Krishnan, G. Sethi, B.B. Aggarwal, Back to basics: how natural
β-boswellic acid in the microtubule assembly dynamics, FEBS Lett. 586 (23) products can provide the basis for new therapeutics, Expert Opin. Investig. Drugs
(2012) 4132–4138. 16 (11) (2007) 1753–1773.
[147] K.-Y. Yoo, S.-Y. Park, Terpenoids as potential anti-Alzheimer’s disease
therapeutics, Molecules 17 (3) (2012) 3524–3538.

11

You might also like