You are on page 1of 17

Neurophysiology, Vol. 51, No.

4, July, 2019

REVIEWS

Altered Neurochemistry in Alzheimer’s Disease:


Targeting Neurotransmitter Receptor Mechanisms
and Therapeutic Strategy
S. Kaur,1 G. DasGupta,2 and S. Singh1
Received January 29, 2019

Alzheimer’s disease (AD) is the most common form of dementia characterized by impaired cognitive
functions associated with altered neurotransmitter levels in the brain. In AD with advancement of age,
symptoms that affect memory, learning ability, language perception, and behavior start to progress.
These shifts correlate with accumulations of amyloid beta plaques and neurofibrillary tangles. Such
pathological changes are thought to distort synaptic neurotransmitter levels and interrupt neuron-to-
neuron functioning. The current drugs in AD, like donepezil, rivastigmine, etc., rely to a significant
extent on ameliorated cholinergic neurotransmission and utilization of anticholinesterase inhibitors.
The recent outbreaks in this disease also target neurotransmitters such as dopamine, serotonin, and
their receptor signaling, rather than focusing on cholinergic neurotransmission. This concept emerges
due to findings of altered neurotransmitter levels in the post mortem brains of AD patients. The current
review summarizes some data underlying mechanisms targeting the role of neurotransmitters and their
association with AD, as well as those related to the respective therapeutic aspects. In addition, the
review describes advances in recent drugs targeting neurotransmission preclinically and clinically for
their neuroprotective role in AD.

Keywords: Alzheimer’s disease, neurotransmitters, receptor modulation, endocannabinoids.

INTRODUCTION gonist (memantine), and this is thought to preserve


cholinergic neurotransmission [2]. Not only
Alzheimer’s disease (AD) is a peculiar neuro­ ACh, but other neurotransmitters, like GABA,
degenerative disorder caused by excessive loss glutamate, and serotonin, also contribute to AD
of neurons, mostly in the hippocampus and pathophysiology. Recently, these neurotransmitters
cerebral cortex. This pathology is characterized are under intense investigation because of their
by cognitive impairment, memory loss, and significant contribution to the learning and memory
behavioral abnormalities. The respective symptoms phenomena [3]. A number of drugs are at present
start to progress with massive loss of cholinergic subjected to preclinical studies and clinical trials for
neurons that synthesize acetylcholine (ACh) and assessing their therapeutic effectiveness in AD [4].
are responsible, to a great extent, for coordinating The hippocampus and neocortex are the major
memory functions [1]. Currently, the available areas associated with learning and memory func­
therapy for AD includes acetylcholinesterase tions, and these have mostly deteriorated in AD.
inhibitors (rivastigmine, galantamine, and done­ The recent outbreaks provide evidence regarding
pezil) and a N-methyl D-aspartate receptor anta­ gradual progression of the disease and loss of
neurons in the cortex, hippocampus, amygdala,
thalamus, and some other subcortical regions of
1
Neuroscience Division, Department of Pharmacology, ISF College of
the brain [5]. However, beyond this, a number of
Pharmacy, Moga, Punjab, India. various inward and outward projections towards the
2
Director ISF College of Pharmacy, Department of Pharmaceutical hippocampal regions emphasize the role of certain
sciences, Moga, Punjab, India.
Correspondence should be addressed to S. Singh
neurotransmitters in the coordination of the memory
(e-mail: shamshersinghbajwa@gmail.com). function [6].

293
0090-2977/19/5104-00293 © 2019 Springer Science+Business Media LLC
294 S. Kaur et al.

Acetylcholine (ACh) in AD. monophosphate (cAMP) [10]. The M 1 ACh recep­


Acetylcholine (ACh) is a neurotransmitter of tors in AD affect hippocampal learning, amy­loid­o ­
the cholinergic system, which plays an essential genesis, and NMDAR-mediated synaptic plasticity,
role in cognitive functions; its level in the brain is as evidenced by various animal knockout transgenic
known to be considerably reduced in AD [7]. The models [11]. The role of M 2 ACh receptors in the
basal forebrain nuclei send significant cholinergic brain is less clear, but this subtype has been reported
projections towards the hippocampus, cerebral to regulate glutamatergic neurotransmission through
cortex, and amygdala [8]. The loss of cholinergic hippocampal neurons toward the prefrontal cortex.
neurons in the basal forebrain and other regions Another study emphasized the presence of M 2
is believed to be one of the crucial reasons for receptors on axonal terminals of glutamatergic
the impairment of memory functions in AD. Post afferents, where they modulate the release of
mortem studies of the brain of AD patients showed glutamate [12]. The possible role of M 2 receptors
a significant reduction in the ACh level; this is seems to be auto-inhibitory in its nature; their
indicative of the strict relation of such shifts to activation inhibits GABAergic signaling and elicits
disorders of cognition and memory. Acetylcholine the release of glutamate. Further, this action disturbs
functions through muscarinic ACh (MACh) and the inhibitory/excitatory balance of receptors
nicotinic ACh (NACh) receptors. Certain evidence and results in the deletion of the M 2 -associated
showed that the marked reduction of ACh receptors CHRM-2 gene, which causes a hypercholinergic
is associated with deposition of amyloid β plaques state in the mammalian brain [13]. In AD, enhanced
and neurofibrillary tangles [9]. The expression of expression of M 2ACh receptors or their long-term
MACh receptors in the brain is well understood, and activation produces a neurotoxic effect in the
recent advancement in their signaling gives reliable brain. Activation of M 3 ACh receptors regulates
therapeutic outcomes in AD (Table 1). the non-amyloidogenic pathway (which is known
The activation of M 1 , M 3 , and M 5 ACh recep­ to be altered in AD), and this further increases
tors increases influx of Ca 2+ and production of accumulation of amyloid β plaques. The reduced
inositol trisphosphate (IP3), whereas M 2 and M 4 expression of these receptors in the hippocampus,
activation leads to inhibition of cyclic adenosine amygdala, and entorhinal cortex affects learning

T a b l e 1.
Receptors Location in the brain Signalling mechanism Functions
• Synaptic plasticity
Hippocampus, • Neuronal excitability,
Subunit α of Gq/11
cerebral cortex, amygdala, • Long-term potentiation
M1 mediates activation of
prefrontal cortex, • Neuronal differentiation during the early development
phospholipase Cβ (PLC)
corpus striatum and thalamus in learning and memory
• Modulation of glutamatergic neurons
Cerebellum, thalamus
Subunit α of Gi and Go
(nucl. basalis, lower levels), Inhibitory modulatory action on dopaminergic
M2 mediates adenylate
hippocampus, amygdala, terminals
cyclase activity
and caudate/putamen
• Regulation of food intake
Cortex, amygdala, Subunit α of Gq/11
• Regulation of learning and memory
M3 hippocampus, olfactory bulb, mediates activation of
• Hypoplasia of the anterior pituitary gland
striatum, thalamus, and pons phospholipase Cβ (PLC)
• Promotion of body growth
• Role in cognition
Hippocampus, dentate gyrus, Subunit α of Gi and Go • Implicated in Psychosis, schizophrenia, and
M4 caudate/putamen, prefrontal mediates adenylate Parkinson’s disease.
cortex, and corpus striatum cyclase activity • Inhibition of the D1 function and suppression of
locomotor activity
Pars compacta
Subunit α of Gq/11
of the substantia nigra, • Facilitates dopamine release (?).
M5 mediates activation
hippocampus, and • Rewarding effect of the abuse drugs.
of phospholipase Cβ (PLC)
ventral tegmental area
Neurotransmitter Alteration and Therapeutic Strategy in AD 295

and leads to declines in memory-associated fear of hippocampal areas. There is, however, one
conditioning [10]. contradictory report showing that amyloid β
Similarly, M 4ACh receptors are allied to striatal provokes activation of β2α4 receptors, as evidenced
glutamatergic projections, hippocampus, and cortex, by cell line studies. This may occur due to increased
where they regulate the cognitive functions. The influx of Ca 2+ , which results in mitochondrial
allosteric modulators of M 4ACh receptors are useful dysfunction and enhanced oxidative stress. One
in the case of cognitive decline and have anti- another recent study reported that amyloid β
psychotic efficacy; these agents became prominent deposition promotes neurotoxicity via inhibition
therapeutic means in AD. Moreover, agonists of of α 7 NACh receptors followed by ERK activation
M 4ACh receptor were shown in various preclinical [17]. Abnormal protein deposition, like amyloid
studies to reduce behavioral disturbances and to β accumulation in the brain, causes dysregulation
improve cognition and conditioned avoidance in the α 7 NACh receptor functioning in AD.
reflexes [11]. Furthermore, M 5 ACh receptors are Additionally, it was reported that direct nicotine
predominantly expressed in the cerebral vasculature administration for 6 months in human subjects
where they regulate cerebral blood flow towards improves memory and cognition deficits, but side
the striatum, cerebral cortex, hippocampus, basal effects, like addiction and adverse effects on the
ganglia, and thalamus. Activation of M 5 ACh cardiovascular system, limit its therapeutic use [16].
receptors enhances striatal dopamine release and Several other molecules targeting nicotinic receptors
also mediates ACh-dependent vasodilation of the are under investigation for the treatment of AD and
cerebral arteries, which was evidenced by genetic are discussed in the management part. In conclusion,
knockout studies [14]. Thus, agents affecting activation of both β 2 α 4 and α 7 NACh receptors
M 5ACh receptors could appear favorable targeted provides noticeable neuroprotective effects in AD
drugs for ischemia-associated AD. (Fig. 1).
Nicotinic receptors are pentameric structural Dopamine in AD.
proteins consisting of five different subunits with Dopamine is synthesized in the midbrain and
a central hydrophilic pore for the flow of cations, known to coordinate a number of functions, like motor
like Na + , Ca 2+ , and K + . Further, these subunits functions, cognition, and reward phenomena [19].
have 16 different small loops formed by α (1-7), A number of studies emphasized that dopaminergic
α (9,10), β (1-4), γ, δ, and ε components. This neurons are lost in AD. Recently, it was revealed [20]
pentameric structure could be homopentameric or that dopaminergic neurons of the ventral tegmental
heteropentameric in its nature; it may be α-bunga­ area (VTA) are significantly involved in cognition,
rotoxin-sensitive or insensitive. The nicotinic receptors formation of memory, and synaptic plasticity.
are widely expressed not only in neuromuscular Degeneration of dopaminergic neurons in the VTA
junctions and autonomic ganglia, but also present occurs in early AD, whereas striatum-associated
in other parts of the brain including neurons and loss of dopaminergic neurons was found to occur in
synapses of hippocampus [15]. Homopentameric α7 later stages. Loss of DA in the striatum contributes
NACh and heteropentameric β2α4 nACh receptors to extrapyramidal symptoms and affects 35-40%
were specifically found to play a vital role in AD. AD patients [21]. Administration of dopaminergic
These subtypes also have a high permeability for supplements, like levodopa or a selegiline-like
Ca 2+, increase glutamatergic neurotransmission, and MAO-B inhibitor, was found to rescue memory
modulate neuronal plasticity via influencing the impairment, as well as neuronal loss in the CA1 region
axonal growth [16]. Activation of α7 nACh and β2α4 of the hippocampus. The favorable neuroprotective
NACh receptors regulates acetylcholine release and effect of dopamine in the hippocampus is considered
inhibits production of inflammatory cytokines, which to be provided by dopaminergic projections targeting
indicates communal contribution of these receptor the hippocampal area of the brain. The cerebral cortex
subtypes to neuroinflammation and cognitive decline. and nucl. accumbens (NAc) are also involved in the
Moreover, these receptors are also involved in regulation of memory and cognition through the
neuronal survival via upregulation of a neurotrophic mesocortico-limbic pathway [20]. The distribution
factor and inhibition of apoptotic factors [17]. of dopaminergic neurons in substantia nigra pars
Oliviero et al. [18] demonstrated that amyloid compacta (SNpc) and VTA helps to understand the
β deposition in synaptosomes induces inhibition role of such units in the coordination of a number of
of β 2 α 4 receptors and decreases the functioning functions in AD.
296 S. Kaur et al.

F i g. 1. Targeting of α7- and α4β2-NACh receptors as a neuroprotective approach.

Ascending projections from the SNpc reach cerebral cortex, and putamen nuclei where they, via
the caudate and putamen nuclei where they the mesostriatal pathway, contribute to the control of
form a mesostriatal pathway and participate in memory, reward-related phenomena, and violation
the regulation of voluntary movements; their [21]. Both above-mentioned projections are
loss produces manifestations of extrapyramidal subjected to degeneration in AD, which determines
symptoms (EPSs) in the late AD stages. Projections the development of the respective associated
originating from the VTA reach the hippocampus, symptoms (Fig. 2).

F i g. 2. The meso-striatal pathway and meso-cortical signalling with the limbic system in AD.
Neurotransmitter Alteration and Therapeutic Strategy in AD 297

Both glutamatergic and cholinergic projections Glutamate in AD.


work in parallel, but overexcitation of glutamatergic Glutamate is an excitatory neurotransmitter
pathways spoils the cholinergic projections. The reported to be involved in the coordination of
evidence regarding decreased release of dopamine, memory, synaptic plasticity, and neurogenesis.
reduction of the number of dopamine receptors, Glutamate not only plays a neuroprotective role
decreased expression of dopamine transporters, and but also is implemented in the excitotoxicity-
decreased activity of tyrosine hydroxylase are well dependent neurotoxic mecha­n isms. The availability
highlighted by available research reports [20, 21]. of L-glutamate in the human brain is approximately
Some statements are related only to the decreased 6–7 μmol/g, which makes this compound functionally
expression of both D1 and D2 receptors in AD, the most abundant free amino acid. There are three
whereas several recent reports emphasize their types of glutamate receptors belonging to the
specific role in AD [22]. ionotropic or metabotropic classes. The ionotropic
Similarly, some studies highlight the role of D1 receptors are further classified as N-methyl-D-
and D2 receptors in cognition via potentiating the aspartate (NMDA)-, α-amino-3-hydroxy-5-methyl-
prefrontal cortex (PFC) signaling. Here, D1-like 4-isoxazolepropionic acid (AMPA)-, and kainate-
receptors are more densified in the prefrontal cortex type receptors. Functionally, these receptors are
(PFC); these receptors are involved in the regulation subclassed as ligand-gated ion channels and are
of working memory, as well as executive functions. of a metabotropic type; the latter belong to the
However, there are less D2 receptors in the PFC GPCR family [23] (Table 2). Both mGluRs1 and
but more in the caudate, VTA, and hippocampal mGluRs5 play crucial roles in the pathology of
regions; they coordinate hippocampus-related AD [24]. The expression of mGluRs1 was found to
cognitive functions. The reduced expression of D2 decrease in AD, as was revealed through available
receptors in AD patients has been reported in the animal studies. Only a few findings were targeted
hippocampus, temporal lobe, and VTA. Not only to activation of mGluRs1 by Aβ aggregation [25].
dopaminergic projections, but also glutamatergic Further, mGluR5 activation leads to Aβ oligomer
ones are present in these regions, and they are also formation. CTEP 2-(chloro-4-((2, 5-dimethyl-1-
affected and regulate excitotoxicity. Perhaps, this is (4-(trifluoromethoxy) phenyl)-1H-imidazol-4-yl)
an evidence that they make D2 receptors protective ethynyl)pyridine) is one of the negative allosteric
against neuroinflammation and excitotoxicity in AD. modulators of mGluRs5; it provides an antagonistic
Here, D2 receptors were shown to be noticeably action on the Aβ pathology in AD. Other studies
responsible for episodic memory and executive showed that treatment with antagonists of mGluRs5,
functions, which are considered extrastriatal effects like MTEP (3-[(2-methyl-1,3-thiazol-4-yl) ethynyl]
in AD. pyridine), provides the neuroprotective effect in AD.

T a b l e 2.
Group Subtype Location Signalling mechanism
Cerebellar cortex, olfactory bulb, lateral septum, globus pallidus,
mGluR1 Couple to Gq/11, stimulate
entopeduncular nucleus, ventral pallidum, and thalamic nuclei
Group I phospholipase C to release Ca2+,
Cerebral cortex, hippocampus, subiculum, olfactory bulb, striatum,
mGluR5 and enhance glutamate release
nucl. accumbens, and lateral septal nucleus
mGluR 2 Cerebellar cortex and olfactory bulb
Negatively regulate adenylyl
Olfactory tubercle, cerebral cortex, dentate gyrus, lateral septal
Group 2 cyclase via coupling to Gαi
mGluR3 nucleus, striatum, nucl. accumbens, amygdaloid nuclei,
and inhibit glutamate release
substantia nigra pars reticulata, and cerebellar cortex
Cerebellum, olfactory bulb, cerebral cortex, hippocampus,
mGluR4 lateral septum, septofimbrial nucleus, striatum,
thalamic nuclei, and spinal dorsal horn Negatively regulate adenylyl
Group 3 mGluR6 Retina cyclase via coupling to Gαi
mGluR7 expressed widely in the brain and inhibit glutamate release
Olfactory bulb, cerebral cortex, and lateral reticular nucleus
mGluR8
of the medulla oblongata
298 S. Kaur et al.

Genetic deletion of mGluR5 in transgenic mice was NMDA receptors occurs in presynaptic, synaptic,
reported to reduce Aβ aggregation [26, 27]. and extrasynaptic regions. Synaptic NMDA receptors
The group-II glutamate receptors exhibit some­ are located on the presynaptic areas of axon
what complex results in AD studies. Normally, terminals; the presynapses are found to be located
activation of mGluRs2 provides neuroprotection 200-300 nm away from the postsynaptic regions.
in this disease, but some contradictory reports The extrasynaptic NMDA (eNMDA) receptors
showed that overactivation of mGluR2 increases are localized on the spine necks, dendritic shafts,
Aβ production [28, 29]. Activation of group-III and somata of the neurons, and their activation
receptors is considered to provide a neuroprotective requires a high amount of glutamate release. The
effect. Similarly, activation of mGluRs7 prevents above receptor type is usually found on tripartite
neuronal degeneration in the basal forebrain synapses (Fig. 3). The tripartite synapse is an area
nuclei but increases glutamate- and Aβ-dependent that not only involves presynaptic and postsynaptic
excitotoxicity damage to these nuclei [30]. neuronal regions, but also is connected to nearby-
Ionotropic receptors, like kainate and AMPA ones, located astrocytes. The synaptic NMDA (sNMDA)
mediate their action through Na + influx, whereas receptors are known to be activated by synaptic
NMDA receptors provide this via Ca 2+ influx. The glutamate discharge and are neuroprotective in
activity of NMDA receptors depends upon Mg 2+ , their nature, as they regulate neuronal survival
which stabilizes them in the normal state, while [32]. In sNMDA receptors, the NR2A subunits are
depolarization prevents these receptors from being activated; such activation further provides anti­
activated. The NMDA receptors consist of three oxidant and antiapoptotic functions. Moreover,
subunits, NR1, NR2 (A, B, C, and D), and NR3 these receptors enhance neuronal survival through
(A and B), which form calcium-permeable ion upregulating the neurotrophic factors. At the same
channels. The NR1, NR2A, and NR2C subunits time, extrasynaptic NMDA (eNMDA)-dependent
are expressed in the temporal cortical regions and activation of NR2B subunits regulates actions
hippocampus within the embryonic stage; they opposite to those of NR2A ones [31]. The NR2B
spread throughout the brain during the neonatal subunits upregulate the excitotoxicity, apoptosis,
development, which is indicative of their role in and neuronal cell death. The eNMDA receptors also
neurogenesis. Similarly, the NR2A and NR2B interact with calpain, upregulate calpain signaling,
subunits are known to play an important role in and promote neurotoxicity, as was evidenced in a
the induction of memory [31]. The distribution of number of animal studies [32].

F i g. 3. NMDA receptor signalling via NR2A neuroprotective and NR2B neurotoxic events in AD.
Neurotransmitter Alteration and Therapeutic Strategy in AD 299

GABA in AD. later on, a decline in such release in age-associated


γ-Aminobutyric acid (GABA) is, probably, the AD was observed. Some research evidence indicated
most important inhibitory neurotransmitter that that Aβ-induced neuronal pore formation enhances
mediates inhibitory signaling through GABA A , calcium release in hippocampal neuronal cultures;
GABA B, and GABA C receptors [33]. The GABA A this leads to enhanced excitotoxicity and, finally,
and GABA C receptors are ligand-gated chloride neuronal death. : During this, there was initial
ion channels, while the GABA B receptors are NMDA activation, which provoked the action on
metabotropic ones, which function via G-protein postsynaptic GABA A receptors. An increase in
coupling. The GABA A receptors consist of two α, GABAergic neurotransmission could interfere with
two β, and one γ subunits and are distributed widely the synaptic function and cause a deficit in long-
in the brain. The GABA C receptors are formed by term potentiation (LTP) in the dentate gyrus, which
α subunits 1-3 and found to be localized in the retina. could be reversed by picrotoxin [34]. On the other
The GABA B receptors are GPC ones functioning hand, hippocampal astrocytes release GABA and
through Gi/Go protein either by inhibiting Ca 2+ glutamate through bestrophin-1(Best-1); these
channels or by opening K + channels [34]. effects per se were observed to be suppressed
The role of GABAergic receptors is implicated in AD [37]. An increased LTP within early AD
in various neurodegenerative disorders, as GABA stages produces cognition deficits via action on the
controls neurotransmission via the respective dentate gyrus. Similarly, an enhanced astrocytic
inhibitory mechanism. Various research studies GABA-dependent LTP deficit in the dentate gyrus
of AD have reported that reduction in the GABA is a possible biomarker of AD [38]. However, a
levels is observed in this case in the temporal, lipoprotein, APOE-4, secreted from GABAergic
parahippocampal, and cingulate cortical regions, neurons disrupts GABAergic neuronal functioning
amygdala, putamen, hypothalamus, and nucl. and impairs cognitive functions [34].
accumbens, [35]. Some studies have also shown An adequate balance between GABAergic and
contradictory results; it was emphasized that GABA glutamatergic neurotransmission is necessary
neurotransmission in the brain is not significantly for normal neuronal functioning. The enhanced
altered in AD [36]. Transgenic mice studies (for excitotoxicity mediates degradation of GABA B
4-month-old tgCRND8 and 18-month-old APP/PS-1 subunits and reduces expression of these receptors
strains, respectively) showed that there was initial in late stages of AD. The GABA B receptors consist
elevation in GABA and glutamate release, while, of GABA B 1 and GABA B 2 subunits that become

F i g. 4. NMDA and GABA signalling in the earlier and late onset of AD.
300 S. Kaur et al.

phosphorylated by the calcium-calmodulin complex that to the dorsal hippocampus caused mild memory
on ser867 and ser783 residues, respectively, impairment [42]. Buspirone is another 5-HT1A
through NMDA activation [39]. This will lead receptor agonist; its administration along with
to endocytosis of GABA B receptors through 8-OH-DPAT enhanced spatial memory better than
lysosoma-mediated degradation observed in both isolated administrations of the above drugs [44]. In
hippocampal and cortical cultured neurons (Fig. 4). another study, parachlorophenylalanine (PCPA) was
However, observations regarding NMDA receptors used, which inhibits tryptophan hydroxylase and
are controversial because activation of synaptic reduces 5-HT synthesis. Administration of PCPA
NMDARs exerts neuroprotective action, while impaired spatial memory, which could be reversed by
extrasynaptic NMDA Rs provide neurotoxic effects. an agonist (like flexinoxan). These studies justified
Synaptic NMDA Rs mediate long-term potentiation the role of 5-HT1A in the formation of spatial
in AD, and their activation increases expression of memory. Activation of 5-HT1B receptors decreased
GABA B Rs that act opposite to the extrasynaptic serotonin release and was associated with changes
NMDA receptors [40]. in emotional memory and some other functions.
Serotonin in AD. It was also reported that 5-HT1B receptors also
Mounting evidence has accumulated indicating affect the release of other neurotransmitters, like
that serotonin regulates, to a significant extent, dopamine and GABA. Post-mortem examinations
mood, emotions, sleep/wake cycle, locomotion, and of AD patients showed that reduced expression of
other behavioral phenomena. Serotoninergic neurons 5-HT1B receptors in the frontal and temporal cortex
localized in the raphe nuclei regulate a variety of correlated with cognitive and behavioral changes.
functions through various serotoninergic receptors. Treatment with selective serotonin reuptake
There are seven different types of serotonin (5-HT) inhibitors (SSRIs), like sertraline, to APP transgenic
receptors present in the brain [41]. The 5-HT1A mice enhanced the mRNA expression of 5-HT1B
receptors have been found to be localized pre- receptors; this was also confirmed in the available
synaptically and post-synaptically throughout the AD studies [45]. The above transgenic study proved
brain. The pathways of these receptors connect that SSRIs could be reliable antidepressants in AD.
the hippocampus with the raphe nuclei and limbic The expression of 5-HT2A receptors was found
structures. Cortical pyramidal neurons, GABAergic to be localized in the neocortex, hippocampus,
neurons, and granular neurons of the hippocampus entorhinal cortex, limbic system, striatum, olfactory
are specifically enriched with 5-HT1A receptors bulb, hypothalamus, amygdala, and basal ganglia.
[42]. Administration of a 5-HT1A receptor agonist Systemic administration of the 5-HT2A selective
modulates theta waves in the hippocampus, and agonist TSB-2 improved exploration of a novel
this fact emphasizes the role of GABAergic septo- object and formation of fear-associated memory in
hippocampal connections in memory formation rats; this is indicative of the role of this receptor
[43]. Activation of 5-HT1A receptors produces type in fear-associated and consolidated memory
hyperpolarization in postsynaptic units and [46]. Another study [47] revealed that activation
decreases cAMP formation, which inhibits calcium of 5-HT2A receptors on hippocampal neurons
channel opening. To understand the role of 5-HT1A modulates glutamatergic neurotransmission for
receptors in learning and memory, various genetic memory consolidation. This effect may arise due to
models were used, and preclinical studies have been increase in the intracellular Ca 2+ level via 5-HT2A
performed. The genetic knockout model with respect receptors, which facilitates the functioning of
to 5-HT1A receptors showed noticeable impairment NMDA receptors and modulates synaptic plasticity.
of spatial learning and memory associated with The decreased expression of 5-HT2A receptors in
hippocampal functions in AD. neocortical (temporal in particular), regions caused
Administration of the HT1A receptor agonist mild cognitive impairment and long-term memory
8-hyd­r oxy-2-(di-n-propylamino)tetralin (8-OH-­ loss [48]. Local injections of amyloid-β in the
DPAT) introduced in different regions of the brain hippocampus reduced the expression of 5-HT2ARs
was shown to improve memory. Similarly, localized in AD model studies [49]. The genetic deletion
administration by intraseptal infusions caused the of 5-HT2B receptors also affected the learning
impairment of memory, while dorsal raphe infusion and memory impairments in the AD mice model
gave no effect on memory. Furthermore, infusion [50]. Likewise, administration of the 5-HT2B/2C
to the medial raphe also improved memory, while receptor antagonist (SB221284) impaired learning
Neurotransmitter Alteration and Therapeutic Strategy in AD 301

and memory, and such effects could be corrected by seems to be interaction of the 5-HT6 receptors with
selective agonists, like fluoxetine and paroxetine Fyn protein kinase, which activates ERK-1/2 and
[51] (Fig. 5). enhances tau phosphorylation in AD [54]. These
The 5-HT4 receptors are specifically localized data are supported by experimental observations;
in the hippocampus; those are associated with a specific agonist of for 5-HT6 receptors,
learning and memory formation [52]. These EMD386088, potentiated cell death in neuronal
receptors regulate the neuronal excitability via cell cultures, while a specific 5-HT6 receptor
changing adenylate cyclase activity, improving antagonist, SB258585, moderated cell death by
synaptic plasticity through the Src/ERK pathway, increasing the intracellular Ca 2+ release [55]. The
and potentiating neurogenesis and neuronal survival 5-HT7 receptors are localized in the hypothalamus,
by modulation of the expression of CREB, Akt, and thalamus, hippocampus, cerebral cortex, and raphe
BDNF in neurons [53]. The 5-HT4 receptors also nuclei. Activation of 5-HT7 receptors results in
reduce amyloid β accumulation through a non- coupling to Gsα, increases the release of сAMP,
amyloidogenic pathway by increasing the activity and leads to increased neuronal excitability [56].
of α-secretase and γ-secretase. Activation of 5-HT4 This mechanism causes pKa activation, which
receptors was reported to regulate the release results in phosphorylation of CREB, in such a way
of ACh, GABA, and dopamine through indirect contributing to memory consolidation. The 5-HT7
mechanisms [52]. receptors are also coupled to G11/12. This activates
Furthermore, available studies indicated that Rho kinase and mitogen-activating protein (MAP)
5-HT6 receptors are involved in memory formation, responsible for neuronal survival and differentiation.
which led us to know their functions in cognition and The Rho protein, through activation of its subunit
memory. These receptors are localized more in the RhoBTB3, prevents degradation of 5-HT3 receptors,
dorsal hippocampus, entorhinal cortex, and striatum which is directly linked to the formation of working
but lesser in the amygdala, substantia nigra, and memory and self-control [57].
several diencephalic nuclei. The 5-HT6 receptors Noradrenaline in AD.
usually couple to Gs and enhance adenylate cyclase In the brain, noradrenergic neurons are prin­
activity, but certain reports indicated that these cipally localized in the locus coeruleus (LC); they
receptors work through Gi and Gq coupling and are mostly responsible for the regulation of fight/
contribute to neurotoxicity. The possible mechanism flight-like reactions, by affecting arousal, stress,

F i g. 5. Signalling mechanisms and roles of 5-HT4, 5-HT6, and 5-HT-7 receptors in AD.
302 S. Kaur et al.

anxiety, executive control, and anger in humans studies, the significant role of NE-ergic LC neurons
[58]. The LC sends its noradrenergic projections in the latter phenomena (memory and cognition) was
towards different regions of the brain, including the emphasized [62, 63].
thalamus, hypothalamus, cerebral cortex, amygdala, DSP-4 (N-(2-chloroethyl)-N-ethyl-2-bromo­
and hippocampus [59]. The LC regulates the release benzylamine hydrochloride) is a selective neurotoxin
of epinephrine, norepinephrine, and dopamine for NE neurons, and its administration in the brain
from presynaptic nerve terminals after binding to leads to destruction of memory [62]. This memory
adrenergic receptors (α, β) for their action. These disturbance gets more severe along with depletion
neurotransmitters are degraded by certain enzymes, of dopamine and acetylcholine. The increase in the
like catechol-O-methyltransferase (COMT) and NE level was shown to improve spatial memory and
monoamine oxidase (MAO), or can be reuptaken recognition memory, which was demonstrated in
via the Na/K pump again to the synapses. Their the radial arm maze test and object recognition test,
activity is upregulated in AD [58]. It was revealed respectively.
that tau-related pathology is associated with The amyloid β processing and synthesis not only
AD-related neurodegeneration in the LC [60]. It was occur in the hippocampus but are also found in
hypothesized in the 1990s that parallel activation of chromaffin cells of catecholaminergic neurons, as
the LC along with that of the peripheral nervous was reported by a few in vitro studies. Stimulation of
system is manifested as increased attention and chromaffin cells by KCl induced the depolarization
vigilance due to the animal’s usual cognition towards enhancement and release of Aβ along with NE [63].
any aversion stimulus [58]. Further, the respective This is explained as follows: these events include
data were supported by the evidence gained from activation of α 2 and β 2 adrenergic receptors present
the Korsakoff ’s syndrome; in this pathology, on synaptic terminals of the LC [64, 65]. There
death of noradrenergic neurons and depletion of are only some studies reporting that an increase
norepinephrine (NE) metabolites in the cerebral in the number of α 2 ARs in the frontal cortex and
fluid are observed. The Korsakoff’s syndrome is a hippocampus is related to polymorphism of the
memory-oriented problem occurring due to chronic gene encoding α 2ARs. These receptors regulate the
consumption of alcohol and malnutrition [61]. This synthesis and production of Aβ, which was shown
example greatly emphasizes the importance of in transgenic studies by interacting with SorLA-like
the NE system in memory and cognition. In a few APP trafficking protein [64]. The β 2ARs are capable

F i g. 6. Involvement of the amygdala and hypothalamus and amyloid protein formation in AD.
Neurotransmitter Alteration and Therapeutic Strategy in AD 303

of directly regulating the activity of the γ-secretase involved in the mediation of a variety of functions
complex and enhancing Aβ production in AD; this like cognition, memory, emotions, and pain
receptor type was found to decrease Aβ production perception. The CB1 receptors are present in the
in transgenic mice [65]. hippocampus, cortex, basal ganglia, and cerebellum.
Remarkably, there is another reliable explanation These receptors were reported to be localized on
regarding damage to the LC in the AD brain. The excitatory and inhibitory presynaptic terminals
LC role in memory formation is regulated by and postsynaptic structures [71]. CB2 receptors are
some projections oriented to the amygdala and rarely found in the healthy brain, but their number is
hypothalamus. The amygdalar projections provide increased in astrocytes and microglia upon damage
intensification of the production of corticotrophin and/or with aging.
releasing factor; the latter acts as a stressor for the The exact role of endocannabinoids signaling in
LC. Similarly, the hypothalamic projections regulate the AD is still under study. It was reported that the
glucocorticoid and hypocretin (orexin) signaling CB1 receptors are either depleted [72] or seemed
that mediate the release of NE in the LC [66, 67] to be unaltered in the hippocampus and cerebral
(Fig. 6). cortex of AD patients [73]. However, the expression
Some evidence indicates that basolateral amyg­ of CB2 receptors in microglia has been reported to
dala-dependent activation of β2ARs regulates the be enhanced at amyloid β aggregation in the brain,
LC functioning under conditions of the formation which may be a protective endogenous phenomenon
of fear-associated memory [66]. The hypothalamic within the initial AD stage [74]. Administration
glucocorticoid and corticosterone projections to the of endogenous endocannabinoids to neuronal
LC regulate memory consolidation in response to cell cultures after exposure to toxic Aβ species
stress. The production of both glucocorticoids and enhances the viability of neurons in in vitro AD
corticosterone is upregulated in stress situations, studies. Simultaneously, exogenous cannabinoids
and this was reported to enhance the amyloid β were shown to increase neuronal survival in AD.
production in AD [67]. Hypothalamus-mediated Results of in vitro and in vivo studies gave reasons
projections regulate orexin signaling and balance to believe that a cannabinoid agonist cannot only
the sleep-wake cycle that is strongly dysregulated in reduce amyloid β aggregation, but also enhance
AD. Administration of an orexin receptor antagonist its clearance through the blood-brain barrier [75,
not only balances the disturbed sleep-wake cycle, 76]. A selective CB1 receptor agonist reduced tau
but also decreases the amyloid β load in AD hyperphosphorylation via decreasing GSK-3β, p-38,
[68, 69]. Thus, neurodegeneration occurring in the and JNK activity [77]. This agent was not only
LC contributes to both cognitive and noncognitive beneficial against specific protein aggregation, but
impairments in AD patients. was also proven to relatively normalize the neuro­
Endocannabinoids in AD. trans­m ission processes in AD. A cannabinoid,
Endocannabinoids are relatively newly iden­tified 9∆-THC, was reported to competitively inhibit
neurohormones synthesized locally and released the acetylcho­linesterase enzyme and enhance cho­
in various regions of the CNS. Unlike most other linergic neurotransmission in AD. A synthetic
neurochemical agents, endocannabinoids belong cannabinoid, HU-211, selectively inhibited NMDA-
to the lipid-mediated eicosanoid family derived induced excitotoxicity in AD [78]. Moreover,
from degradation of phospholipids. The most numerous studies indicated that cannabinoid
common endogenous cannabinoids are arachi­­do­n o­ signaling is protective against neuroinflammation,
ylethanolamine (AEA, or anandamide), 2-arachi­ mitochondrial damage, oxidative stress, and
donoylglycerol (2-AG), and others (2-arachi­ neurodegeneration in this pathology. Similarly,
donylglyceryl ether (2-AGE), virodhamine, and cannabinoid signaling in GABAergic neurons
N-arachi­d onyldopamine). The discovery of endocan­ protects the brain against a cognitive decline by
nabinoids is attributed to the ancient use of the regulating long-term potentiation-related memory
Cannabis plant, which retains specific actions on the [70].
CNS mostly due to the presence of a psychoactive Drugs Targeting Neurotransmitters in the
constituent named 9∆-tetrahydrocannabinol (9∆- Preclinical and Clinical Phases of AD.
THC) [70]. Endocannabinoid receptors (CB-1 and To bridge neurochemistry-targeting drugs in
CB-2 Rs) are widely distributed in the CNS and AD, we examined various clinical and preclinical
304 S. Kaur et al.

study reports. A recent study [79] reported that like 77-LH-28-1, VU0357017, and VU364572,
about 23% of the drugs targeting neurotransmitters were reported to have a better M 1 selectivity and
have been tested in AD-related clinical trials up to permeability in the brain [80]. Most M1 agonists failed
2017. This number is the highest among the other to demonstrate sufficient success during clinical trials,
probable mechanisms related to the treatment of and this led to the focusing attention on nicotinic
this pathology (like anti-tau therapy, anti-amyloid β agonists like, EVP-6124. These agents demonstrated
therapy, and metabolic drugs) [79]. The summarized reliable efficacy in AD studies [80]. Excitatory
data include not only drugs reaching the stage of neurotransmission studies have already focused
immediate clinical trials, but also neurotransmitter- on memantine-like drugs, whose efficacy has been
targeting drugs used in preclinical studies (Fig. 7). clinically approved. The other targeting molecules
The list of the preclinically tested compounds include MK-801(an NMDA-blocking agent),
is too long and wide, because a great number of huperzine-A, neramexane (an uncompetitive NMDA
mo­ l ecules are being evaluated for their better antagonist), ifenprodil (a GluN2B antagonist), and RO
efficacy as neuroprotective means in AD (Fig. 8). 25-6981 (a GluN2B antagonist); these drugs showed a
Upregulation of the cholinergic system by reliable efficacy over excitatory neurotransmission in
flavonoids and carbamate derivatives is still AD studies [31]. Inhibitory neurotransmission plays
under investigation through in silico, in vitro, and a controversial role in AD because both “agonist”
in vivo studies. Flavonoids, like rutin, myricetin, and “antagonist” studies reported in the literature
quercetin, kaemferol, and macluraxanthone, were gave rather contradictory results. The GABA A ago­
evaluated for their effectiveness in inhibiting the nists (etazolate, muscimol, propofol, MRK-016,
acetylcholinesterase enzyme in AD. Agonists of M1 CGS9896, Ro-4938581, and Ro-4882224) and
muscarinic receptors, like AF102B, AF267B, AF150, GABA B antagonists (SGS742 and CGP55845) were
and AF292B, were shown to be useful neuroprotective reported to improve cognition and memory in the
agents in various AD studies. Other molecules, respective studies [34].

F i g. 7. Drugs targeting different neurotransmitters, which have been tested in clinical evaluation as therapeutic means in AD.
Neurotransmitter Alteration and Therapeutic Strategy in AD 305

One recent report [20] revealed the role of the spatial deficits induced by LC degeneration
dopamine in the modulation of hippocampal memory and was shown to be protective over NA neuronal
and synaptic plasticity through administration loss in AD. A number of drugs, like vindeburnol,
of dopamine supplements (like levodopa) and allopregnenolone, sertraline (SSRI), venlaflaxine
monoamine oxidase-B inhibitors (like selegiline). (SNRI), desipramine, and atomoxetine (NARI),
The expression of MAO mRNA was found to were implemented in various AD studies and have
increase in the brain of AD patients. Polymorphism shown some good results for neuroprotection and
of certain MAO-related genes was also reported over cognition loss [81]. Recently, cannabinoids
in the brain of such patients. Earlier, it was shown have attracted much greater attention in the
that MAO-A activity is upregulated in AD, but the search of therapeutic agents in AD. Endogenous
interaction of MAO-B with presenilin emphasizes cannabinoids (dronabinol, AEA, 2-AG, and noladin
the role of the latter enzyme subtype in amyloid β ether) added to amyloid β-treated cells enhanced
processing. Administration of an MAO-B inhibitor, neuronal survival and decreased amyloid β overload
selegiline, helped to reverse the amyloid β pathology in in vitro studies [77]. Similarly, arachidonyl-2-
and cognition loss in transgenic animal studies. chloroethylamide (ACEA), CB2 selective agonists
Catechol-O-methyltransferase (COMT) is another (JWH-015 and JWH-133), and mixed CB1/CB2
enzyme responsible for dopamine degradation, and receptor agonists (9-THC, HU-210, and WIN55,
its expression was found to increase after brain 212-2) were shown to be neuroprotective agents
injury, trauma, and lipopolysacharide injections; in various AD studies [82]. HU-210 is a synthetic
so, the role of the above enzyme in AD-related cannabinoid capable of reducing excitotoxicity via
machinery cannot be ruled out. Another drug is direct binding to NMDA receptors, which makes it
L-DOPS (droxidopa), which is expected to reverse a reliable agent in AD research [78].

F i g. 8. Drugs targeting different neurotransmitters, which have been tested in preclinical evaluation as therapeutic means in AD.
306 S. Kaur et al.

Thus, it should be recognized that there are 4. A. Kumar, A. Singh, and Ekavali, “A review on Alz­
numbers of loopholes regarding the pathogenesis heimer’s disease pathophysiology and its management:
an update,” Pharmacol. Rep., 67, No. 2, 195–203 (2015).
of AD. In the current review, the role of various
5. H. Braak and K. Del Tredici, “The preclinical phase
neurotransmitters has been discussed on the of the pathological process underlying sporadic Alz­
basis of their receptor mechanisms behind their heimer ’s disease,” Brain, 138, No. 10, 2814–2833
contribution in pathophysiological signaling in (2015).
AD. Targeting the cholinergic system could be a 6. M. Llorens-Martín, L. Blazquez-Llorca, R. Benavides-
rational approach to halt AD development. At the Piccione, et al., “Selective alterations of neurons and
circuits related to early memory loss in Alzheimer’s
same time, other neurochemicals, like dopamine,
disease,” Front. Neuroanat., 8, 1–38 (2014).
GABA, glutamate, and endocannabinoids, are 7. A. Contestabile, “The history of the cholinergic hypo­
involved in the modulation of memory, cognition, thesis,” Behav. Brain Res., 221, No. 2, 334–340 (2011).
and neurodegeneration in AD and its models; thus, 8. E. C. Ballinger, M. Ananth, D. A. Talmage, and
different aspects of the respective phenomena L. W. Role, “Basal forebrain cholinergic circuits and
should be examined. There is a need to look at the signaling in cognition and cognitive decline,” Neuron,
91, No. 6, 1199–1218 (2016).
possible hidden mechanisms and multidrug therapy
9. H. T. Ferreira-Vieira, M. I. Guimaraes, R. F. Silva, and
for AD treatment. Further studies are required to M. F. Ribeiro, “Alzheimer’s disease: targeting the chol­i ­n­-
explore the immune- and genetic-based alterations ergic system,” Curr. Neuropharmacol., 14, No. 1, 101–
in the neurotransmitter levels in AD patients. So, 115 (2016).
to overcome these neurological problems, targeting 10. T. Ahmed, S. Zahid, A. Mahboob, and S. M. Far­
neurotransmitters, agents suppressing hyper- hat, “Cholinergic system and post-translational modi­
fications: an insight on the role in Alzheimer’s disease,”
immune responses, and those altering enzyme
Curr. Neuropharmacol., 15, No. 4, 480–494 (2017).
levels may be useful to devise superior remedies, 11. D. J. Foster, D. L. Choi, P. J. Conn, and J. M. Rook,
which could minimize symptoms and elucidate AD “Acti­v ation of M1 and M4 muscarinic receptors as pote­
pathophysiology. n­tial treatments for Alzheimer’s disease and schizo­
phrenia,” Neuropsychiatr. Dis. Treat., 10, 183–191
Acknowledgment. We are highly thankful to the chairman (2014).
of the ISF College of Pharmacy, Moga, Punjab, India, for 12. J. D. Guo, R. Hazra, J. Dabrowska, et al., “Presynap­tic
providing a favorable educational and research environment. mus­c arinic M 2 receptors modulate glutamater­­gic tran­
smission in the bed nucleus of the stria terminalis,”
This paper is a review of the available literature; so, Neuropharmacology, 62, No. 4, 1671–1683 (2012).
special confirmation of its correspondence to the existing 13. A. S. Gibbons, W. J. Jeon, E. Scarr, and B. Dean, “Chan­
ethical standards for the experimental studies is not ges in muscarinic M2 receptor levels in the cortex of
necessary. subjects with bipolar disorder and major depressive
disorder and in rats after treatment with mood stabilizers
The authors, S. Kaur, G. DasGupta, and S. Singh, declare and antidepressants,” Int. J. Neuropsychopharmacol.,
the absence of any conflict in commercial or financial 19, No. 4, 1–9 (2015).
relations, relationships with organizations or persons 14. E. P. Lebois, C. Thorn, J. R. Edgerton, et al., “Muscarinic
that in any way could be related to the study, and also in receptor subtype distribution in the central nervous
interrelations of the co-authors. system and relevance to aging and Alzheimer’s disease,”
Neuropharmacology, 136, Pt. C, 362–373 (2017).
15. E. K. Lebbe, S. Peigneur, I. Wijesekara, and J. Tytgat,
“Conotoxins targeting nicotinic acetylcholine receptors:
an overview,” Mar. Drugs, 12, No. 5, 2970–3004 (2014).
REFERENCES 16. S. Lombardo and U. Maskos, “Role of the nicotinic
acetylcholine receptor in Alzheimer’s disease pathology
1. H. Jahn, “Memory loss in Alzheimer ’s disease,” and treatment,” Neuropharmacology, 96, Pt. B, 255–262
Dialogues Clin. Neurosci., 15, No. 4, 445–454 (2013). (2015).
2. N. Guzior, A. Wieckowska, D. Panek, and B. Ma­- 17. K. T. Dineley, A. A. Pandya, and J. L. Yakel, “Nicotinic
lawska, “Recent development of multifunctional agents ACh receptors as therapeutic targets in CNS disorders,”
as po­t ential drug candidates for the treatment of Trends Pharmacol. Sci., 36, No. 2, 96–108 (2015).
Alzheimer’s disease,” Curr. Med. Chem., 22, No. 3, 373– 18. G. Olivero, M. Grilli, J. Chen, et al., “Effects of soluble
404 (2015). β-amyloid on the release of neurotransmitters from rat
3. D. S. Strac, D. Muck-Seler, and N. Pivac, “Neuro­t rans­ brain synaptosomes,” Front. Aging Neurosci., 6, 166,
mitter measures in the cerebrospinal fluid of patients doi: 10.3389/fnagi.2014.00166 (2014).
with Alzheimer’s disease: a review,” Psychiatr. Danub., 19. C. Rangel-Barajas, I. Coronel, and B. Florán, “Dopamine
27, No. 1, 14–24 (2015). receptors and neurodegeneration,” Aging Dis., 6, No. 5,
349–368 (2015).
Neurotransmitter Alteration and Therapeutic Strategy in AD 307

20. A. Nobili, E. C. Latagliata, M. T. Viscomi, et al., 34. Y. Li, H. Sun, Z. Chen, et al., “Implications of GABA­
“Dopamine neuronal loss contributes to memory and ergic neurotransmission in Alzheimer’s disease,” Front.
reward dysfunction in a model of Alzheimer’s disease,” Aging Neurosci., 8, 1–12 (2016).
Nat. Commun., 8, 14727, doi: 10.1038/ncomms14727 35. K. Govindpani, B. Calvo-Flores Guzmán, C. Vinnakota,
(2017). et al., “Towards a better understanding of GABAergic
21. A. Martorana and G. Koch, “Is dopamine involved in remodeling in Alzheimer’s disease,” Int. J. Mol. Sci., 18,
Alzheimer’s disease?” Front. Aging Neurosci., 6, 252– No. 8, 1–41 (2017).
257 (2014). 36. A. Kwakowsky, B. Calvo‐Flores Guzmán, M. Pandya, et
22. L. Nyberg, N. Karalija, A. Salami, et al., “Dopamine D2 al., “GABA A receptor subunit expression changes in the
receptor availability is linked to hippocampal–caudate human Alzheimer’s disease: hippocampus, subiculum,
functional connectivity and episodic memory,” Proc. entorhinal cortex and superior temporal gyrus,” J.
Natl. Acad. Sci. USA, 113, No. 28, 7918–7923 (2016). Neurochem., 145, No. 5, 374–392 (2018).
23. J. Lewerenz and P. Maher, “Chronic glutamate toxicity 37. L. P. Diniz, V. Tortelli, I. Matias, et al., “Astrocyte
in neurodegenerative diseases – what is the evidence?” trans­forming growth factor beta 1 protects synapses
Front. Neurosci., 9, 1–20 (2015). against Aβ oligomers in Alzheimer’s disease model,” J.
24. T. M. Sanderson, E. L. Hogg, G. L. Collingridge, and Neurosci., 37, No. 28, 6797–6809 (2017).
S. A. Corrêa, “Hippocampal metabotropic glutamate 38. Z. Wu, Z. Guo, M. Gearing, and G. Chen, “Tonic inhi­
receptor long-term depression in health and disease: bition in dentate gyrus impairs long-term potentiation
focus on mitogen activated protein kinase pathways,” J. and memory in an Alzheimer’s disease model,” Nat.
Neurochem., 139, Suppl. 2, 200–214 (2016). Commun., 5, 1–25 (2014).
25. S. V. Ovsepian, L. Blazquez-Llorca, S. V. Freitag, et al., 39. S. Kantamneni, “Cross-talk and regulation between
“Ambient glutamate promotes paroxysmal hyperacti­ glutamate and GABA B receptors,” Front. Cell. Neurosci.,
vity in cortical pyramidal neurons at amyloid plaques 9, 1–7 (2015).
via presynaptic mGluR1 receptors,” Cereb. Cortex, 27, 40. C. Tackenberg, S. Grinschgl, A. Trutzel, et al., “NMDA
No. 10, 4733–4749 (2016). receptor subunit composition determines beta-amyloid-
26. A. Hamilton, M. Vasefi, C. Vander Tuin, et al., “Chronic induced neurodegeneration and synaptic loss,” Cell
pharmacological mGluR5 inhibition prevents cognitive Death Dis., 4, 1–10 (2013).
impairment and reduces pathogenesis in an Alzheimer 41. J. Masson, M. B. Emerit, M. Hamon, and M. Dar­m on,
disease mouse model,” Cell Rep., 15, No. 9, 1859–1865 “Serotonergic signaling: multiple effectors and pleio­
(2016). tropic effects,” Wiley Interdisciplin. Rev., Membrane
27. A. Hamilton, G. W. Zamponi, and S. S. Ferguson, “Glu­ Transport and Signaling, 1, No. 6, 685–713 (2012).
tamate receptors function as scaffolds for the regulation 42. Y. Glikmann-Johnston, M. M. Saling, D. C. Reutens, and
of β-amyloid and cellular prion protein signaling J. C. Stout, “Hippocampal 5-HT1A receptor and spatial
complexes,” Mol. Brain, 8, No. 1, 1–9 (2015). learning and memory,” Front. Pharmacol., 6, 289–299
28. S. H. Kim, J. W. Steele, S. W. Lee, et al., “Proneurogenic (2015).
group II mGluR antagonist improves learning and 43. C. M. Monaco, K. M. Gebhardt, S. M. Chlebowski,
reduces anxiety in Alzheimer Aβ oligomer mouse,” Mol. et al., “A combined therapeutic regimen of buspiro­n e
Psychiatry, 19, No. 11, 1235–1242 (2014). and environmental enrichment is more efficacious than
29. F. Caraci, G. Molinaro, G. Battaglia, et al., “Targeting either alone in enhancing spatial learning in brain-
group II metabotropic glutamate (mGlu) receptors for injured pediatric rats,” J. Neurotrauma, 31, No. 23,
the treatment of psychosis associated with Alzheimer’s 1934–1941 (2014).
disease: selective activation of mGlu2 receptors ampli­ 44. R. W. Welford, M. Vercauteren, A. Trébaul, et al., “Sero­
fies β-amyloid toxicity in cultured neurons, whereas tonin biosynthesis as a predictive marker of serotonin
dual activation of mGlu2 and mGlu3 receptors is neuro­ pharmacodynamics and disease-induced dysregulation,”
protective,” Mol. Pharmacol., 79, No. 3, 618–626 (2011). Sci. Rep., 6, 1–10 (2016).
30. Z. Gu, J. Cheng, P. Zhong, et al., “Aβ selectively 45. W. Tajeddinn, T. Persson, S. Maioli, et al., “5-HT1B and
impairs mGluR7 modulation of NMDA signaling in other related serotonergic proteins are altered in APPswe
basal forebrain cholinergic neurons: implication in mutation,” Neurosci. Lett., 594, 137–143 (2015).
Alzheimer’s disease,” J. Neurosci., 34, No. 41, 13614– 46. W. Tajeddinn, T. Persson, J. Calvo-Garrido, et al., “Phar­
13628 (2014). macological modulations of the serotonergic system in
31. Y. Zhang, P. Li, J. Feng, and M. Wu, “Dysfunction of a cell-model of familial Alzheimer’s disease” J. Alz­
NMDA receptors in Alzheimer’s disease,” Neurol. Sci., heimers Dis., 53, No. 1, 349–361 (2016).
37, No. 7, 1039–1047 (2015). 47. G. Zhang and R. W. Stackman, Jr, “The role of serotonin
32. C. C. Rudy, H. C. Hunsberger, D. S. Weitzner, and 5-HT2A receptors in memory and cognition,” Front.
M. N. Re­e d, “The role of the tripartite glutamatergic Pharmacol., 6, 1–17 (2015).
synapse in the pathophysiology of Alzheimer’s disea­s e,” 48. G. Zhang, H. N. Ásgeirsdóttir, S. J. Cohen, et al., “Sti­
Aging Dis., 6, No. 2, 131–148 (2015). mulation of serotonin 2A receptors facilitates conso­
33. C. Wu and D. Sun, “GABA receptors in brain deve­ lidation and extinction of fear memory in C57BL/6J
lopment, function, and injury,” Metab. Brain Dis., 30, mice,” Neuropharmacology, 64, No. 1, 403–413 (2013).
No. 2, 367–379 (2015).
308 S. Kaur et al.

49. L. Marner, G. M. Knudsen, K. Madsen, et al., “The 64. Y. Chen, Y. Peng, P. Che, et al., “Alpha(2A) adrenergic
reduction of baseline serotonin 2A receptors in mild receptor promotes amyloidogenesis through disrupting
cognitive impairment is stable at two-year follow-up,” APP-SorLA interaction,” Proc. Natl. Acad. Sci. USA,
J. Alzheimers Dis., 23, No. 3, 453–459 (2011). 111, No. 48, 17296–17301 (2014).
50. P. M. Pitychoutis, A. Belmer, I. Moutkine, et al., “Mice 65. A. Thathiah, K. Horré, A. Snellinx, et al., “Beta-arrestin 2
lacking the serotonin HTr 2B receptor gene present an regulates A beta generation and gamma secretases
antipsychotic-sensitive schizophrenic-like phenotype,” activity in Alzheimer’s disease,” Nat. Med., 19, No. 1,
Neuropsychopharmacology, 40, No. 12, 2764–2773 43–49 (2013).
(2015). 66. A. Uematsu, B. Z. Tan, and J. P. Johansen, “Projection
51. M. E. Gibbs and L. Hertz, “Serotonin mediation of spe­c ificity in heterogeneous locus coeruleus cell
early memory formation via 5-HT2B receptor-induced po­p ulations: implications for learning and memory,”
glycogenolysis in the day-old chick,” Front. Pharmacol., Learn. Mem., 22, No. 9, 444–451 (2015).
5, 54–65 (2014). 67. D. A. Bangasser, K. R. Wiersielis, and S. Khantsis, “Sex
52. H. Hagena and D. Manahan-Vaughan, “The seroton­- differences in the locus coeruleus-norepinephrine system
er­g ic 5-HT4 receptor: A unique modulator of hippocam­ and its regulation by stress,” Brain Res., 1641, Pt. B,
pal synaptic information processing and cognition,” 177–188 (2016).
Neurobiol. Learn. Mem., 138, 145–153 (2017). 68. J. H. Roh, H. Jiang, M. B. Finn, et al., “Potential role
53. J. Pascual-Brazo, E. Castro, Á. Díaz, et al., “Modu­la­ of orexin and sleep modulation in the pathogenesis of
tion of neuroplasticity pathways and antidepres­s ant- Alzheimer’s disease,” J. Exp. Med., 211, No. 13, 2487–
like behavioral responses following the short-term 2496 (2014).
(3 and 7 days) administration of the 5-HT4 receptor 69. Y. E. Ju, B. P. Lucey, and D. M. Holtzman, “Sleep
agonist RS67333,” Int. J. Neuropsychopharmacol., 15, and Alzheimer disease pathology- a bidirectional
No. 5, 631–­6 43 (2011). relationship,” Nat. Rev. Neurol., 10, No. 2, 115–119
54. M. J. Ramírez, “5-HT 6 receptors and Alzheimer ’s (2014).
disease,” Alzheimers Res. Ther., 5, No. 2, 1–15 (2013). 70. G. Bedse, A. Romano, A. M. Lavecchia, et al., “The
55. H. M. Yun and H. Rhim, “5-HT 6 receptor ligands, role of endocannabinoids signaling in the molecular
EMD386088 and SB258585, differentially regulate mechanisms of neurodegeneration in Alzheimer ’s
5-HT6 receptor-independent events,” Toxicol. In Vitro, disease” J. Alzheimers Dis., 43, No. 4, 1115–1136
25, No. 8, 2035–2040 (2011). (2015).
56. O. Stiedl, E. Pappa, Å. Konradsson-Geuken, and 71. V. Di Marzo, N. Stella, and A. Zimmer, “Endo­c anna­
S. O. Ög­r en, “The role of the serotonin receptor sub­ binoid signaling and the deteriorating brain,” Nat. Rev.
types 5-HT 1A and 5-HT 7 and its interaction in emotional Neurosci., 16, No. 1, 30–42 (2015).
learning and memory,” Front. Pharmacol., 6, 1–17 72. M. Solas, P. T. Francis, R. Franco, and M. J. Ramírez,
(2015). “CB2 receptor and amyloid pathology in frontal cortex
57. N. Zareifopoulos and C. Papatheodoropoulos, “Effects of Alzheimer’s disease patients,” Neurobiol. Aging, 34,
of 5-HT-7 receptor ligands on memory and cognition,” No. 3, 805–808 (2013).
Neurobiol. Learn. Mem., 136, 204–209 (2016). 73. R. Ahmad, K. Goffin, J. Van den Stock, et al., “In vivo
58. S. R. Chamberlain and T. W. Robbins, “Noradrenergic type 1 cannabinoid receptor availability in Alzheimer’s
modulation of cognition: therapeutic implications,” J. disease,” Eur. Neuropsychopharmacol., 24, No. 2, 242–
Psychopharmacol., 27, No. 8, 694–718 (2013). 250 (2014).
59. G. Aston-Jones and B. Waterhouse, “Locus coeruleus: 74. A. V. Savonenko, T. Melnikova, Y. Wang, et al.,
from global projection system to adaptive regulation of “Can­n a­b inoid CB2 receptors in a mouse model of
behavior,” Brain Res., 1645, 75–78 (2016). Aβ amyloidosis: immunohistochemical analysis and
60. H. Braak and K. Del Tredici, “Where, when, and in what suitability as a PET biomarker of neuroinflammation,”
form does sporadic Alzheimer’s disease begin?” Curr. PLoS One, 10, No. 6, 1–18 (2015).
Opin. Neurol., 25, No. 6, 708–714 (2012). 75. E. Janefjord, J. L. Mååg, B. S. Harvey, and S. D. Smid,
61. R. P. Vetreno, R. L. Ramos, S. Anzalone, and L. M. Sa­- “Cannabinoid effects on β amyloid fibril and aggregate
vage, “Brain and behavioral pathology in an animal formation, neuronal and microglial-activated neuro­to­
model of Wernicke’s encephalopathy and Wernicke– xicity in vitro,” Cell. Mol. Neurobiol., 34, No. 1, 31–42
Korsakoff syndrome,” Brain Res., 1436, 178–192 (2013).
(2012). 76. A. M. Martín-Moreno, B. Brera, C. Spuch, et al., “Pro­
62. S. Rajabi, A. Shamsizadeh, H. Amini, et al., “Effect of longed oral cannabinoid administration prevents neuro­
DSP-4 induced central noradrenergic depletion on tactile inflammation, lowers β-amyloid levels and improves
learning in rat,” Neurol. Res., 34, No. 1, 80–84 (2012). cognitive performance in Tg APP 2576 mice,” J.
63. T. Toneff, L. Funkelstein, C. Mosier, et al., “Beta Neuroinflammation, 9, 1–8 (2012).
amyloid peptides undergo regulated co-secretion with 77. E. Aso, S. Juvés, R. Maldonado, and I. Ferrer, “CB2
neuropeptide and catecholamine neurotransmitters,” cannabinoid receptor agonist ameliorates Alzheimer-like
Peptides, 46, 1–22 (2013). phenotype in AβPP/PS1 mice,” J. Alzheimers Dis., 35,
No. 4, 847–858 (2013).
Neurotransmitter Alteration and Therapeutic Strategy in AD 309

78. I. Ferrer, “Cannabinoids for treatment of Alzheimer’s 81. D. L. Feinstein, S. Kalinin, and D. Braun, “Causes, con­
disease: moving toward the clinic,” Front. Pharmacol., se­q uences, and cures for neuroinflammation mediated
5, 1–37 (2014). via the locus coeruleus: noradrenergic signaling system,”
79. J. Cummings, G. Lee, T. Mortsdorf, et al., “Alzheimer’s J. Neurochem., 139, Suppl. 2, 154–178 (2016).
disease drug development pipeline: 2017,” Alzheimers 82. E. Aso and I. Ferrer, “CB2 cannabinoid receptor as
Dement. (NY), 3, No. 3, 367–384 (2017). potential target against Alzheimer’s disease,” Front.
80. A. Kumar, C. M. Nisha, C. Silakari, et al., “Current and Neurosci., 10, 243–253 (2016).
novel therapeutic molecules and targets in Alzheimer’s
disease,” J. Formos. Med. Assoc., 115, No. 1, 3–10
(2016).

You might also like