Available online at www.sciencedirect.com

ScienceDirect

The role of GPCRs in neurodegenerative diseases:

avenues for therapeutic intervention

1 2 1,2,3,4

Yunhong Huang , Nicholas Todd and Amantha Thathiah

Neurodegenerative diseases represent a large group of cognitive function, memory loss, and negative personality

neurological disorders with heterogeneous clinical and changes [3–5]. The pathological features of AD include

pathological profiles. The majority of current therapeutic the accumulation of amyloid b (Ab) in amyloid plaques

strategies provide temporary symptomatic relief but do not and hyperphosphorylated aggregates of the microtubule-

target the underlying disease pathobiology and thus do not associated tau in neurofibrillary tangles, which are

affect disease progression. G protein-coupled receptors first detected in the frontal and temporal lobes and then

(GPCRs) are among the most successful targets for therapeutic slowly progress to the other areas of the neocortex [5].

development of central nervous system (CNS) disorders. Many VaD is the second most common cause of dementia with a

current clinical therapeutic agents act by targeting this class of variable age of onset. VaD patients display a disturbance in

receptors and downstream signaling pathways. Here, we frontal executive function [6] and multiple cerebrovascular

review evidence that perturbation of GPCR function pathologies, including vessel occlusion, arteriosclerosis,

contributes to the pathophysiology of various hypertensive, aneurysms, and various forms of arteritis

 

neurodegenerative diseases, including Alzheimer’s disease, [7 ,8 ]. Frontotemporal dementia (FTD) is a major cause

Frontotemporal dementia, Vascular dementia, Parkinson’s of dementia in persons under the age of 65 [9] and

disease, and Huntington’s disease. is characterized by neuropsychiatric symptoms and

behavioral, motor, and cognitive impairments [10]. The

Addresses pathological features of FTD include the abnormal depo-

1

Department of Neurobiology, University of Pittsburgh School of sition of three major —tau, transactive response

Medicine, 3501 Fifth Avenue, BST3, Pittsburgh, PA 15213, USA DNA-binding protein 43 (TDP-43), and fused in sarcoma

2

University of Pittsburgh Brain Institute, University of Pittsburgh School

(FUS) protein [10] in brain regions such as the hippocam-

of Medicine, 3501 Fifth Avenue, BST3, Pittsburgh, PA 15213, USA

3 pus, frontal cortex, and striatum [11].

Pittsburgh Institute for Neurodegenerative Diseases, University of

Pittsburgh School of Medicine, 3501 Fifth Avenue, BST3, Pittsburgh,

PA 15213, USA Parkinson’s disease (PD) is second most common neuro-

4

KU Leuven Center for Human Genetics, Leuven 3000, Belgium

degenerative disease, with an average onset of 50–60

years of age [12]. PD is characterized by motor and

Corresponding author: Thathiah, Amantha ([email protected])

non-motor symptoms. The prominent motor symptoms

in PD patients include bradykinesia, rigidity, tremor, and

Current Opinion in Pharmacology 2017, 32:96–110 gait disorders [13]. Non-motor clinical features include

This review comes from a themed issue on Neurosciences cognitive impairment and neuropsychiatric symptoms

[13]. The pathological features of PD include deposi-

Edited by David Chatenet and Terence E. He´ bert

tion of Lewy bodies and abnormal aggregates of the

For a complete overview see the Issue and the Editorial

a-synuclein protein in several brain regions, such as the

Available online 10th March 2017

substantia nigra and temporal cortex, and the loss of

http://dx.doi.org/10.1016/j.coph.2017.02.001 dopaminergic neurons in the substantia nigra [13].

1471-4892/ã 2017 Elsevier Ltd. All rights reserved.

Similar to PD, Huntington’s disease (HD) patients suf-

fer from motor and non-motor symptoms. HD patients

suffer from motor symptoms such as chorea, bradykine-

sia, impaired coordination, and rigidity and non-motor

symptoms such as depression and slowed cognitive

Introduction function [14]. HD is caused by a CAG trinucleotide

Neurodegenerative diseases are a major cause of disabil- repeat expansion in the Huntingtin (Htt) [14].

ity and premature death among the elderly people world- The CAG repeats vary from 6 to 35 nucleotides in

wide [1]. Alzheimer’s disease (AD), Vascular dementia unaffected individuals. A longer series of CAG repeats

(VaD), Frontotemporal dementia (FTD), Parkinson’s (>36) are present in HD patients and inversely correlate

disease (PD), and Huntington’s disease (HD) are the with the age of onset [15]. The deposition of HTT is

among the most prevalent neurodegenerative diseases most frequent in the cerebral cortex, and much less in

[2]. AD is the most common neurodegenerative disease other brain regions such as striatum, hippocampus, and

and the predominant cause of dementia in the population cerebellum [16]. Collectively, AD, VaD, FTD, PD,

over 65 years of age. AD is characterized by impaired and HD are neurodegenerative diseases with clinical

Current Opinion in Pharmacology 2017, 32:96–110 www.sciencedirect.com

Role of GPCRs in neurodegenerative diseases Huang, Todd and Thathiah 97

features that include cognitive deficits, motor impair- function [28]. Acetylcholinesterase inhibitors and mem-

ments, and neuropsychiatric symptoms. antine are the only available symptomatic treatments that

slow the decline in cognitive function in individuals with

G protein-coupled receptors (GPCRs) have been impli- AD [24]. In this section, we highlight some of the GPCRs

cated in the pathogenesis of several neurodegenerative that have been rigorously evaluated in the modulation

diseases [17,18], including AD, VaD, FTD, PD, and HD. of cognitive function in AD mouse models in recent

GPCRs are the largest family of membrane proteins [19]. literature. Additional GPCRs that have been implicated

Over 370 non-sensory GPCRs have been identified of in the pathophysiology of AD have been included in

which more than 90% are expressed in the brain, where Table 1.

they play important roles in mood, appetite, pain, vision,

immune regulation, cognition, and synaptic transmission Glutamate receptors mediate most of the excitatory neu-

[20]. GPCR ligands include a variety of molecules such as rotransmission in the mammalian brain [29]. The meta-

photons, ions, biogenic amines, peptide, hormones, botropic (mGluR) family mediate

growth factors, and lipids [21]. Consequently, GPCRs glutamate neurotransmission. MGluR5 has been shown

represent the most common target for therapeutic drugs. to be involved in cognitive function and Ab generation.

Here, we mainly focus on the GPCRs that have been Genetic deletion of mGluR5 has been shown to alleviate

reported in the past 5 years and several well-documented cognitive impairment and Ab production in an APPswe/

GPCRs that have been reported to be involved in the PSEN1DE9 AD mouse model, which overexpresses

pathophysiology of the neurodegenerative diseases human APP harboring the Swedish mutation and human

mentioned above. We review the correlation between presenilin 1 lacking exon 9 [30]. Interestingly, pharmaco-

changes in GPCR expression and/or activity with the logical inhibition of mGluR5 with 3-[(2-methyl-1,3-thia-

neuropathological hallmarks and clinical symptoms zol-4-yl)ethynyl]-pyridine (MTEP), an antagonist, has

of these neurodegenerative diseases and discuss the also been shown to alleviate the cognitive deficits in

currently available therapeutic strategies targeting the the same AD mouse model [31]. Similarly, treatment

GPCRs discussed in the text. with the mGluR5 negative allosteric modulator 2-

chloro-4-((2,5-dimethyl-1-(4-(trifluoromethoxy)phenyl)-

Alzheimer’s disease 1H-imidazol-4-yl)ethynyl) pyridine (CTEP) alleviates

GPCRs and cognitive deficits in AD the cognitive deficits and reduces the amyloid plaque

AD leads to significant degeneration of various brain burden in two AD mouse models [32]. These studies

regions and the alteration of multiple neurochemical suggest that allosteric modulators of mGluR5 may be an

pathways. Magnetic resonance imaging (MRI) studies effective therapeutic strategy for some AD cases.

have shown that a reduction in the volume of the hippo-

campus and entorhinal cortex, which are affected early in Extensive serotonergic denervation of the neocortex and

disease progression [5,22], and cortical thickness of the hippocampus has been observed in AD patients. Reduc-

medial temporal, inferior temporal, temporal pole, angu- tion in 5-hydroxytryptamine (5-HT, serotonin) and 5-

lar gyrus, superior parietal, superior frontal, and inferior HT1A, 5-HT2A, 5-HT4, and 5-HT6 receptor levels have

frontal cortex correlate with the cognitive deficits been reported in the hippocampus and/or prefrontal

observed in AD patients [3] (Figure 1a). Furthermore, cortex of AD patients. In rodent models, activation of

changes in multiple neurochemical pathways, including 5-HT2A and 5-HT4 receptors leads to an improvement in

the acetylcholine, serotonin, adenosine pathways have hippocampal-dependent learning and memory [33,34] via

been shown to be involved in the cognitive impairments G protein- or b-arrestin-dependent activation of extracel-

observed in AD. lular signal-regulated kinase (ERK) [35,36]. In contrast,

antagonism of the 5-HT1A and the least studied 5-HT5A

Currently, there is no effective treatment for AD. Levels receptors has been shown to ameliorate the memory

of acetylcholine are reduced in the brains of AD patients deficits in a rat AD model [37,38], possibly through an

[23]. As such, acetylcholinesterase inhibitors have been inhibition of Gi signaling and activation of protein kinase

shown to temporarily ameliorate disease symptoms [24] A (PKA), which leads to the activation of the NMDA

by decreasing acetylcholine breakdown, which results in receptor [39,40]. Interestingly, both 5-HT6 receptor ago-

an increase in cholinergic neurotransmission and a mild nists and antagonists enhance learning and memory [41]

improvement in cognitive function. Excitotoxicity due through potentially different mechanisms of action. Acti-

to overstimulation of glutamatergic neurotransmission vation of the 5-HT6 receptor has been shown to stimulate

[25] is also associated with the pathophysiology of AD Gs protein-dependent brain-derived neurotrophic factor

[26]. Memantine is an N-methyl-D-aspartate (NMDA) (BDNF) mRNA expression and Fyn kinase-dependent

receptor antagonist that inhibits NMDA receptor-medi- activation of ERK1/2 in wild-type rats [42]. Both BDNF

ated calcium influx into neurons [27] and protects exces- and ERK1/2 have been shown to be associated with

sive glutamate-induced neuronal death and excitotoxicity cognitive function [43,44]. In contrast, 5-HT6 receptor

[26], providing temporary improvement in cognitive antagonists have been shown to stimulate glutamate and

www.sciencedirect.com Current Opinion in Pharmacology 2017, 32:96–110 98 Neurosciences

Figure 1

(a) Alzheimer’s Disease PR IPL

PC

MTG EC HP ITG TP H P

(b) Frontotemporal Dementia

S ACC STG MPC

A OFC MTG

TP

(c) Vascular Dementia

DPC DPC

HP ITG MTG

Current Opinion in Pharmacology

Schematic representation of the association between brain atrophy and the clinical symptoms observed in Alzheimer’s disease, Frontotemporal

dementia, and Vascular dementia. Blue indicates brain regions that undergo atrophy and are associated with cognitive deficits. Green indicates

Current Opinion in Pharmacology 2017, 32:96–110 www.sciencedirect.com

Role of GPCRs in neurodegenerative diseases Huang, Todd and Thathiah 99

acetylcholine release in rat brains, which has been shown of Crhr1 in the PSAPP AD mouse model, which over-

to improve scopolamine- and MK-801-induced deficits in express a chimeric mouse/human APP gene with human

associative learning [42]. These studies support the APP Swedish mutation and human presenilin 1 lacking



potential benefit of selective modulation of the 5-HT exon 9, leads to a reduction in amyloid pathology [59 ].

receptor subtypes for AD therapy. Pharmacological studies in the Tg2576 AD mouse model,

which overexpresses human APP with the Swedish muta-

Expression of the adenosine A1 and A2A receptors (A1R tion, with the CRHR1 antagonist antalarmin in acutely (7-

and A2AR) has been reported to be elevated in the frontal days) or in chronically (9-months) stressed mice reduces

cortex of the human AD brain [45]. Caffeine, a nonselec- Ab production and involves the Gs signaling pathway

tive AR inhibitor, has been shown to enhance memory [60]; however, pre-treatment with antalarmin failed to

consolidation in humans [46] and reduce Ab levels and inhibit an increase in Ab levels in acutely (3-hours)

improve cognitive function in an AD mouse model [47]. stressed wild-type mice. In vitro cell-free g-secretase

Similarly, caffeine and the A2AR antagonist SCH58261 activity assays with the CRHR1 antagonists astressin,

has been shown to be protective against Ab-induced antalarmin, and NBI-27914 have been shown to modulate

cognitive impairment [48]. Interestingly, conditional Ab generation in the absence of CRHR1, suggesting that

deletion of astrocytic A2ARs has been shown to enhance the compounds tested may have CRHR1-independent



alleviate the memory deficits in AD transgenic mice effects on the modulation of g-secretase activity [57 ].



through Gs-coupled signaling [49 ], whereas activation Treatment of wild-type mice with the CRHR1 antagonist

of the Gi-coupled A1R and inhibition of PKA has been antalarmin reduces depression-like behaviors, whereas

shown enhance long-term depression (LTD) [50]. These genetic deficiency of Crhr2 leads to an increase in depres-

studies potentially suggest that activation of Gs-coupled sion-like behaviors [61]. Although both receptors have

receptors, such as the A2AR, which activates PKA, may considerable sequence similarity, the two receptors have

suppress LTD and promote long-term plasticity (LTP), different expression patterns in the brain and affinities for

whereas Gi-coupled receptors, such as the A1R may be CRH [62]. Interestingly, CRHR1 is more abundantly

involved in the induction of LTD. expressed in the pituitary gland, and atrophy of this

region is associated with the neuropsychiatric symptoms

In addition to GPCRs with identified ligands, the orphan in AD [63]. The in vivo studies suggest that a highly

GPCR GPR3 has been shown to modulate Ab generation selective antagonist specific for CRHR1 may be benefi-

and cognitive function in vivo. Levels of GPR3 are cial for the symptoms of depression in AD; however,

elevated in the human AD brain [51,52]. Genetic deletion careful monitoring of Ab levels would also be necessary

of Gpr3 has been shown to alleviate the learning and to fully assess the therapeutic potential.

memory deficits in an AD mouse model and reduce



amyloid pathology in four AD mouse models [53 ].

The GPR3-mediated effect on amyloid pathology Frontotemporal dementia

involves b-arrestin recruitment, independently of Gs- FTD is a heterogeneous neurodegenerative disease

coupling [51]. A more comprehensive discussion on the caused by degeneration and atrophy of the frontal and

GPCRs involved in the pathogenesis of AD is the subject temporal lobes. In general, FTD encompasses a wide

of recent reviews [54,55]. Together with the GPCRs such range of neuropathologies associated with mutations in

as 5-HT receptors, adenosine receptors that are involved several , including tau, TDP-43, and FUS, which

in affected neurochemical pathways in AD suggest viable leads to deterioration in behavior, personality, and motor

therapeutic avenues for the treatment of cognitive deficits functions [10]. MRI and single-photon emission comput-

in AD. erized tomography (SPECT) reveal abnormalities and

atrophy in the frontal and temporal lobes of FTD

patients. Further post-mortem examination of FTD

GPCRs and neuropsychiatric symptoms in AD patient brains shows additional degeneration of the stria-

The corticotrophin-releasing hormone (CRH) receptor tum [64] (Figure 1b). FTD patients present with a variety

1 and 2 (CRHR1 and CRHR2) are GPCRs associated of neuropsychiatric, behavioral, motor, and cognitive



with depression [56,57 ]. Interestingly, a greater density impairments [64,65] including decline in social skills,

of amyloid plaques has been observed in the hippocam- depression, compulsive behavior, agitation, bradykinesia,

pus of AD patients with a previous history of major and/or apathy. Symptom heterogeneity has led to multi-

depression [58]. Reports also show that genetic deletion ple diagnostic clinical categories such as behavioral

(Figure 1 Legend Continued) brain regions that undergo atrophy and are associated with neuropsychiatric symptoms. Orange indicates brain

regions that undergo atrophy and are associated with motor impairments. Abbreviations for the indicated brain regions include: amygdala (A),

anterior cingulate cortex (ACC), dorsolateral prefrontal cortex (DPC), entorhinal cortex (EC), hypothalamus (H), hippocampus (HP), inferior parietal

lobule (IPL), inferior temporal gyrus (ITG), medial prefrontal cortex (MPC), medial temporal gyrus (MTG), orbitofrontal cortex (OFC), pituitary (P),

posterior cingulate (PC), precuneus (PR), striatum (S), superior temporal gyrus (STG), and temporal pole (TP).

www.sciencedirect.com Current Opinion in Pharmacology 2017, 32:96–110 100 Neurosciences ] ]  ] ] ] ]  ] 135,155 135 ] 136 141–143 144,145,151,152 154 137,140 137 R[ 2A A D1R [ M4R [ s Motor impairments a ] a ]  ein-coupled receptor 52; GPR55, G 117 118 R, 5-hydroxytryptamine receptor 4; 5- 4 ] CB1R [ ] GPR52 [ ne receptor M4; mGluR2, metabotropic glutamate R, cysteinyl 1; D1R, dopamine 1 a ] ] D1R [ ] mGluR5 [ 157,158 ] D2R [ ] D2R [ ] mGluR2 [ 129,131 130 , sphingosine 1-phosphate receptor. 1 131 127 13 122 123 receptor; BAI1, brain-specific angiogenesis inhibitor 1; CB1R, R[ 2A 2A mGluR4 [ M4R [ ] GPR37] [ GPR55 [ ,153 ,153   ]A ] D1R [ ] M1R [ 101 101 R, adenosine A 106 106 ] D2R [ 2A  R1 [ R2 [ R[ R[ R, 5-hydroxytryptamine receptor 2C; 5-HT B B 94,95 98 1A 2A 2C 5-HT 5-HT D1R [ GABA GABA M1R [ receptor; A a a 1 ] ] 10,86,87,150 10,86,87,150 R, adenosine A 1 -Aminobutyric acid B receptor; GIPR, glucose-dependent insulinotropic polypeptide receptor; GPR3, G protein- D1R [ D2R [ g R, B a a ] R, 5-hydroxytryptamine receptor 2A; 5-HT ] a a 2A ] ]   a 89 73,74 72–74 ] 79 71 R[ R[ R[ 1A 2A 2C 5-HT 5-HT R, 5-hydroxytryptamine receptor 6; A 6 ] 5-HT a a ] ] 149 61 61 R[ 2A R, 5-hydroxytryptamine receptor 1A; 5-HT CRHR1 [ 1A 5-HT a ] ] ] a a a ]] 5-HT ] mGluR5 [ ] ] ]  ] ]] CRHR2 [ OXTR [ a a a ] 159 161 ] ] 37 34 38 ] ] ]  30 33 41 163 164 ] R[ R[ 53 R[ R[ R[ 162 167 1 48,49 160 156 B 166 165 R[ R[ [ 48 1A 2A 4 5A 6 R, 5-hydroxytryptamine receptor 5A; 5-HT 1 R[ 5A R[ Studies which were not conducted in animal disease models. 1 2A

A BAI1 [ 5-HT 5-HT HT D1 receptor; D2R, dopamine D2 receptor; DOR, delta-; GABA 5-HT cannabinoid type 1 receptor; CRHR1, corticotrophin-releasing hormone receptor 1; CRHR2, corticotrophin-releasing hormone receptor 2; CysLT 5-HT CysLT coupled receptor 3; GPR30, G protein-coupled receptor 30;receptor GPR37, 2; G mGluR4, protein-coupled glutamate 37; receptor GPR48, 4; G mGluR5, protein-coupled metabotropic receptor glutamate 48; receptor GPR52, 5; G OXTR, prot ; S1P protein-coupled receptor 55; M1R, muscarinic M1; M3R, muscarinic acetylcholine receptor M3; M4R, muscarinic acetylcholi M3R [ GPR30 [ GABA DOR [ Abbreviations: Table 1 GPCRs that have been studied in humansAD and/or animal models Cognitive deficits Neuropsychiatric5-HT symptoms Neuropsychiatric symptoms Motor impairments Cognitive deficits Motor impairments Cognitive deficit GIPR [ GPR3 [ GPR48 [ M1R [ mGluR5 [ S1P FTDa VaD PD HD A

Current Opinion in Pharmacology 2017, 32:96–110 www.sciencedirect.com

Role of GPCRs in neurodegenerative diseases Huang, Todd and Thathiah 101

variant FTD (bvFTD), primary progressive aphasia cortex and isocortical brain regions of bvFTD patients,

(PPA), semantic dementia (SD), and FTD with parkin- which may precede the neurodegeneration observed in

sonism-17 (FTDP-17) [10]. FTD variants affect both select frontotemporal brain regions. Electrophysiology

distinct and overlapping brain regions and multiple neu- and behavioral studies suggest that mGluR5 activation

rochemical pathways, which poses a significant challenge enhances NMDA receptor function, whereas mGluR5

for studying and treating the disease. Here, we discuss the inhibition exacerbates the effects of NMDA receptor

GPCRs that are the most prominent candidates for ther- blockade [80,81]. These studies suggest that mGluR5

apeutic treatment of the neuropsychiatric and motor may be in involved in reduced NMDA receptor neuro-

impairments in FTD. A more comprehensive list of transmission. Interestingly, mGluR5 has also been shown

the GPCRs involved in FTD can be found in Table 1. to play a role in Ab generation, memory, locomotor

function, and anxiety in an AD mouse model, indicating

GPCRs and neuropsychiatric symptoms in FTD the multiple functions of the receptor [30].

Deficits in emotion recognition skills are thought to

contribute to deficits in empathy and inappropriate social

behavior in FTD. Several studies suggest that the neuro- GPCRs and motor impairments in FTD

peptide oxytocin is an important mediator of social behav- The dopamine D1 and D2 receptors (D1R and D2R)

ior and neuropsychiatric behaviors in patients with FTD have been reported to play an important role in FTD.

[66]. In mammals, oxytocin is primarily produced within Both the D1R and D2R are most abundantly expressed in

the hypothalamic brain regions and is shuttled to the the striatum [82]. Because of disease heterogeneity, both

pituitary for systemic release or projected to various brain DR antagonists (antipsychotics) and agonists, which pre-

regions for paracrine signaling of the oxytocin receptor dominantly target the D2R, have been used to treat FTD.

(OXTR) in brain regions such as the amygdala and Clinically, DR antagonists are used to treat behavioral

anterior cingulate cortex, which have been implicated symptoms such as agitation and disinhibition, and DR

in the pathophysiology of FTD [67]. agonists are used to treat motor symptoms such as

rigidity and bradykinesia in bvFTD and FTDP-17,

Oxytocin administration has been shown to potentially respectively [10]. Typical antipsychotics such as halo-

improve social interactions [67] and facilitate the devel- peridol and fluphenazine are not commonly used to treat

opment of GABAergic synapses, which inhibit signals FTD patients due to neuroleptic side-effects associated

that lead to fear and anxiety [68–70]. Interestingly, intra- with the high D2R affinity [83]. In contrast, antipsycho-

nasal administration of oxytocin to FTD patients leads to tics such as olanzapine, quetiapine, and risperidone also

improved social interactions, namely patient–caregiver have a high affinity for the D2R, but rapidly dissociate,

interactions [71]. resulting in fewer side effects [84]. Dopamine dysfunc-

tion has also been reported to be involved in behavioral

Levels of the 5-HT1A and 5-HT2A receptors are reduced deficits in HD (see below); however, decreased D2R

in the anterior cingulate cortex [72] and orbitofrontal and levels has been reported to be the cause of these impair-

medial prefrontal cortex of FTD patients [73,74] and the ments [85].

frontal and temporal cortex of bvFTD patients [72–74].

Clinically, selective serotonin reuptake inhibitors (SSRIs) FTDP-17 patients who experience rigidity and brady-

such as fluoxetine, fluvoxamine, and sertraline, which kinesia primarily display a decrease in presynaptic dopa-

increase 5-HT levels by blocking 5-HT reuptake, have minergic nerve terminals and postsynaptic D2R binding

been used to provide symptomatic relief for depression in the striatum. Consequently, FTDP-17 patients are

and repetitive or compulsive behaviors observed in currently treated with DR agonists such as carbidopa

patients with multiple variants of FTD [75]. Interest- and levodopa, which have been approved for the treat-

ingly, lower levels of the 5-HT1A and 5-HT2A receptors in ment of PD [10,86,87] despite potential exacerbation of

the hippocampus and prefrontal cortex of AD patients the behavioral and psychotic symptoms. Apathy has also

have been shown to lead to cognitive deficits in contrast to been associated with reduced dopaminergic activity

the neuropsychiatric symptoms observed in FTD [76]. [88]. Interestingly, a randomized controlled trial with

Collectively, these studies indicate that 5-HT1A and 5- the 5-HT2C antagonist agomelatine has shown promis-

HT2A receptors are involved in the pathophysiology of ing results with improvement in apathy and an indirect

both AD and FTD. increase in prefrontal dopaminergic tone in FTD



patients [89 ]. Clinical trials of agomelatine in AD

mGluR5 and the NMDA receptors are co-localized in and PD patients also indicate a reduction in apathy

cortical brain regions and act in a cooperative fashion and depression in AD patients [90] and a significant

[77,78]. Specifically, mGluR5 is involved in the induction decrease in depression and motor symptoms of PD [91],

of NMDA receptor-dependent forms of synaptic plastic- indicating the involvement and beneficial effects of



ity and excitotoxicity [78]. Leuzy et al. [79 ] recently targeting the 5-HT2C receptor in three neurodegenera-

showed a decrease in mGluR5 availability in paralimbic tive disorders.

www.sciencedirect.com Current Opinion in Pharmacology 2017, 32:96–110

102 Neurosciences

Vascular dementia serotonergic system as a therapeutic target in both VaD

VaD is the second most common cause of dementia and is and AD.

associated with multiple cerebrovascular pathologies

 

[7 ,8 ]. MRI studies reveal cortical and subcortical micro- Clinical trials of FDA approved AD drugs such as done-

infarcts and atrophy of the frontal and temporal lobes, pezil, an acetylcholinesterase inhibitor, galantamine, a



hippocampus, and striatum of VAD patients [7 ,92] nicotinic acetylcholine receptor agonist, and memantine,

(Figure 1c). The M1 muscarinic acetylcholine receptor a NMDAR antagonist, have been conducted with VaD

(M1R) has been shown to be involved in cognitive patients with some positive cognitive outcomes [107,108];

function [93]. In this regard, hippocampal damage caused however, GPCRs have not been extensively studied in

by cerebrovascular occlusion leads to a reduction in the VaD patients but provide additional neurochemical tar-

3

number of M1Rs and reduced [ H]quinuclidinyl benzi- gets for therapeutic intervention in VaD.

late binding to all muscarinic acetylcholine receptors in

the hippocampus of a chronic cerebral hypoperfusion Parkinson’s disease

(CCH) rat model of VaD [94,95]. In addition, D1Rs have GPCRs and cognitive deficits in PD

been shown to elicit long-term potentiation and enhance PD is a neurodegenerative disease with clinical features

memory storage in the hippocampus [96,97]. A reduction that include motor and non-motor symptoms [13,109].

in D1Rs has been reported in CCH rats. Agonist-induced Approximately 25% of individuals with PD develop mild

activation of the D1R in the dentate gyrus (DG) attenu- cognitive impairment (MCI) [110], including attention,



ates the cognitive impairments in CCH rats [98 ]. These executive function, episodic memory, visuoperceptual/



studies indicate that both M1Rs and D1Rs are involved in visuospatial function, and language deficits [111 ]. A 20-

cognitive function in CCH VaD models. year follow-up study indicates that approximately 80% of

PD patients develop dementia (PDD) [112]. Patholog-

GABAB receptors in the DG regulate synaptic plasticity, ically, MRI studies of PD patients with MCI have

learning, and memory [99]. Lower levels of GABABR1 reduced volume of the nucleus accumbens (NAc)

 

and GABABR2 have been reported in the hippocampus of [113,114 ], thalamus [113], and amydala [114 ] relative

CCH rats. Administration of the GABABR agonist baclo- to cognitively normal individuals with PD (Figure 2a);

fen to CCH rats leads to an increase in GABABR expres- however, changes in the volume of the thalamus and the

sion and an improvement in spatial learning and memory amydala appear to be PD cohort-dependent.

[100]. In contrast, increased GABABR activity in the

hippocampus of CCH rats has also been observed, and The dopamine D1 and D2 receptors (D1R and D2R) are

treatment with the GABABR antagonist saclofen has been highly expressed in multiple brain regions, including the



shown to improve spatial learning and memory [101 ]. striatum, NAc, and substantia nigra [115]. Levels of both

receptors are elevated in PD patients and are associated

Interestingly, combinatorial treatment with acamprosate, with the development of dopamine denervation super-

which reduces glutamatergic neurotransmission, and the sensitivity [116]. Infusion of the D1R partial agonist SKF

GABABR agonist baclofen has been shown to regulate the 38393, but not the D2R agonist quinpirole, into the NAc

balance between excitatory and inhibitory neuronal sig- of wildtype rats enhanced the accuracy of visuospatial

naling, protecting against Ab-induced neurotoxicty and discrimination [117], whereas treatment with a D1R

alleviating cognitive deficits in an AD mouse model antagonist SCH 23390 decreased accuracy [117], indicat-



[102 ]. The drug PXT864, a combination of baclofen ing that the D1R is involved in visuospatial function.

and acamprosate, is currently in the phase II clinial trials Treatment with the D2R antagonist sulpiride or D2R

for the treatment of AD [103]. These reports suggest the knockdown in the NAc reduced attention performance or



GABABR plays an important role in cognitive function induced attention impairment [118 ], respectively, which

and that further study of the GABABRs will be necessary suggests that the D2R is involved in the regulation of

to delineate the role of these GPCRs in VaD. attention. Taken together, these studies provide evi-

dence to support a role for the D1R and the D2R involved

5-HTRs are abundant in the frontal and temporal cortices in visuospatial and attentional dysfunction in PD-MCI,

[75,104,105] and have been shown to play an important respectively.

3

role in cognition and memory formation. Increased [( H)-

3

WAY 100635] and [( H)-ketanserin] radioligand binding GPCRs and motor impairments in PD

has been reported in post-mortem brain samples from Dopamine deficiency within the basal ganglia leads to

patients to 5-HT1A and 5-HT2A receptors, respectively, parkinsonian motor symptoms, including bradykinesia,

possibly due to decreased 5-HT availability. Additionally, muscular rigidity, rest tremor, and postural and gait

5-HT1A receptors binding positively correlates with pre- impairment [13]. MRI studies have shown that degener-

served cognition based on the mini-mental state exam ation of the putamen nucleus, which is part of the striatum

[106]. Reduced 5-HT1A receptors in AD brains leads to and basal ganglia, correlate with the motor deficits

cognitive impairments [76], further highlighting the observed in PD [119]. Currently, dopamine replacement

Current Opinion in Pharmacology 2017, 32:96–110 www.sciencedirect.com

Role of GPCRs in neurodegenerative diseases Huang, Todd and Thathiah 103

Figure 2

(a) Parkinson’s Disease

T

P

NA A

(b) Huntington’s Disease

C P

Current Opinion in Pharmacology

Schematic representation of the association between brain atrophy and clinical symptoms observed in Parkinson’s disease and Huntington’s

disease, both of which display motor impairments. Blue indicates brain regions that undergo atrophy and are associated with cognitive deficits.

Orange indicates brain regions that undergo atrophy and are associated with motor impairments. Abbreviations for the indicated brain regions

include: amygdala (A), caudate (C), nucleus accumbens (NA), putamen (P), thalamus (T).

therapy with levodopa (L-dopa), a chemical precursor of have been developed to reduce the adverse effects asso-

dopamine, is the most effective drug for the symptomatic ciated with L-dopa therapy [121]. The agonists display

treatment of PD [120]. However, higher doses of L-dopa better pharmacokinetic and pharmacodynamic properties

are required to compensate a decline in clinical efficacy than L-dopa with reduced incidence or delayed onset of

after long-term L-dopa therapy, which results in adverse dyskinesia [121]. Currently, the D1R agonist rotigotine

effects, such as motor fluctuations and motor complica- and the D2R agonists bromocriptine and lisuride have

tions such as dyskinesia [120]. Several D1R and D2R been used as a monotherapy or an adjunctive therapy to

agonists, including rotigotine, bromocriptine and lisuride, L-dopa for the treatment of PD motor symptoms

www.sciencedirect.com Current Opinion in Pharmacology 2017, 32:96–110

104 Neurosciences

[122,123]. However, similar to L-dopa, dopamine agonist Chronic glutamate-mediated excitotoxicity has been sug-

therapy also leads to a decline in efficacy with long-term gested to contribute to disease progression [132].

treatment, limiting the use of dopaminergic therapy. A mGluRs, including mGluR2 and mGluR5, are widely

promising non-dopaminergic alternative is the A2AR expressed in the brain in the neocortical layers, hippo-

antagonist istradefylline, which was recently approved campus, striatum, thalamus/hypothalamus, and cerebel-

in Japan as a combination therapy with L-DOPA to treat lum. Activation of presynaptic mGluR2 [133] and block-

motor dysfunction in PD without increasing the risk of ade of postsynaptic mGluR5 [134] inhibit glutamate



dyskinesia [124 ,125]. These studies suggest that a com- release and prevent excitotoxicity. Treatment of R6/2

bination therapy could be an alternative approach for the HD transgenic mice, which express the N-terminally

treatment of parkinsonian motor symptoms. truncated human HTT with 141–157 CAG repeats,

with either the mGluR2 agonist LY379268 or the

A balance between the dopaminergic and cholinergic mGluR5 antagonist 2-methyl-6-(phenylethynyl)-pyri-

system is important in PD [126]. Reduced striatal dine (MPEP) leads to a reduction in hyperactivity

dopamine in PD leads to overactivity of cholinergic [135]. MPEP treatment also reduces the decline in

interneurons and excess acetylcholine release in the motor coordination [135]. Consistent with these findings,

Q111/Q111

striatum [126]. Anticholinergics such as trihexyphenidyl genetic deletion of mGluR5 in Hdh knock-in

and biperiden, which are selective for the M1R, are mice, which express a 109 CAG repeat insertion, leads to

effective in reducing tremors in PD patients [13]. an improvement in motor coordination and a reduction

Anticholinergics show little effect on bradykinesia in HTT aggregation [135]. These studies suggests that

and rigidity, suggesting a specific role for M1Rs in mGluRs regulate motor function and HTT protein

PD-associated tremor. Genetic deletion of the musca- aggregation in HD.

rinic acetylcholine receptor 4 (M4R) in mice reduces

antipsychotic-induced catalepsy [127], a PD motor Accumulation of the mutant HTT protein is considered

symptom, supporting a role for the muscarinic acetyl- to initiate the cytotoxicity which leads to HD. A recent

choline receptors in PD motor symptoms. Conversely, study reported that knockdown of the orphan GPCR

dopamine agonists, used to treat motor symptoms, may GPR52, which is highly expressed in the striatum, re-

Q140/Q140

worsen cognition in PD patients, thereby complicating duces HTT protein levels in the striatum of Hdh

therapeutic options in patients suffering with PD with mice by promoting HTT clearance and suppresses HD

dementia (PDD) [128]. phenotypes in both patient-induced pluripotent stem

cell (iPSC)-derived neurons and in a Drosophila HD



Two orphan GPCRs, GPR37 and GPR55, have also been model [136 ], suggesting that striatal degradation of

implicated in motor coordination [129,130]. Interestingly, mutant HTT requires the GPR52-mediated upregulation

in a drug-induced parkinsonian tremor model, genetic of cyclic adenosine monophosphate (cAMP) levels.

deletion of Gpr37 leads to an attenuation of tremulous jaw

movements (TJMs) in response to the nonselective mus- In both humans and HD animal models, D1R and D2R

carinic acetylcholine receptor agonist pilocarpine [131]. expression is reduced in early and late stage HD [137]. In

Treatment with the A2AR antagonist SCH-58261 also early stage HD, patients experience hyperkinesia, poten-

attenuates pilocarpine-induced TJMs [131], an effect tially due to hyperactivity of the dopamine pathway [138].

which is not observed in GPR37-deficient mice. Collec- In contrast to GPR52, activation of the D2R, which

tively, these studies suggest that strategies aimed at the interacts with Gai and negatively regulates cAMP levels,

two orphan GPCRs may represent an alternative thera- has been shown to lead to an increase in HTT aggregation

peutic avenue for intervention in PD. [138] whereas inhibition of the D2R with the antagonist

haloperidol has been shown to reduce HTT aggregation

and protect against striatal cell death, which may be

Huntington’s disease beneficial in the early stages of HD [139].

HD is a progressive neurodegenerative disorder that

presents clinically with involuntary movements, impaired D1R antagonists have also been studied in HD. The D1R

coordination, depression, and slowed cognitive function. antagonist SCH23390 has been shown to prevent dopa-

HD is caused by a CAG trinucleotide repeat expansion in mine- and glutamate-induced cell death in YAC128 mice

the first exon of the Huntingtin (Htt) gene. The CAG [140], which express multiple copies of the full-length

repeats vary from 6 to 35 nucleotides in unaffected human mutant HTT protein with 128 glutamine repeats,

individuals. A longer series of CAG repeats (>36) are mainly composed of CAG repeats and nine interspersed

present in HD patients and inversely correlate with the CAA repeats. These studies suggest that GPR52-, and

age of onset [15]. Structural MRI studies indicate exten- D1R-/D2R-specific signaling regulate HTT degradation

sive degeneration of the striatum and, to a lesser extent, and aggregation, respectively, and may serve as potential

the globus pallidus, thalamus, and hippocampus in HD therapeutic targets for HD drug discovery. These studies

patients [14] (Figure 2b). also suggest that striatal-enriched modulators of HTT

Current Opinion in Pharmacology 2017, 32:96–110 www.sciencedirect.com

Role of GPCRs in neurodegenerative diseases Huang, Todd and Thathiah 105

levels may contribute to the selective vulnerability of These GPCRs represent potential opportunities for the

striatal neurons. Further studies will be required to deter- development of disease-modifying therapies. Given the

mine whether the D1Rs, D2Rs, and GPR52 are involved symptom heterogeneity of and the variety of GPCRs

in the motor impairments observed in HD patients. implicated in disease progression of different neurode-

generative disease, a combinatorial therapeutic approach,

The cannabinoid type 1 receptor (CB1R) has been shown targeting multiple GPCRs, may prove to be beneficial to

to mitigate HTT aggregation in the R6/2 HD mouse slow and perhaps halt disease progression. In this regard,

model. The CB1R is normally highly expressed at syn- inhibition of both the A2AR and CRHR1 may improve

apses in the neocortex, hippocampus, and basal ganglia cognitive function and reduce depression in AD patients.

[141]; however, CB1R levels are reduced in R6/2 mice A combination of of OXTR and DR agonists in FTD

[142]. Moreover, chronic treatment of R6/2 mice with the patients may alleviate the neuropsychiatric and motor

9

CB1R agonist D -tetrahydrocannabiol (THC) in R6/2 symptoms. Furthermore, mechanistic studies to under-

alleviated motor symptoms relative to vehicle-treated stand the interaction between different neurochemical

animals [142]. In PC12 cells expressing a mutation in pathways are critical to reduce potential side-effects

the HTT [143], activation of the CB1R, which couples to associated with monotherapies. As such, the combination

Gai, with the CB1R agonists HU210 or WIN55212-2 has therapy of L-DOPA and the A2AR antagonist istradefyl-

been shown to alleviate the cell death associated with line, which target two neurochemical pathways, have

HTT aggregation. Taken together, these studies high- reduced side-effects relative to L-DOPA monotherapy

light CB1R agonists as potential therapeutics for HD and in PD patients. Abnormal accumulation of the proteins

suggest a complex role for cAMP in HTT aggregation and mentioned in this review also leads to various pathological

degradation. changes in the brain, including mitochondrial dysfunc-

tion, oxidative stress, and neuroinflammation. Therefore,

The A2AR has also been proposed to be a therapeutic an alternate avenue for therapeutic intervention is to

target for HD. A2ARs are localized throughout the brain target the GPCRs involved in neuroprotection. In this

but are primarily found in medium spiny neurons in the regard, neuropeptides acting on GPCRs, such as vasoac-

striatum [144]. Presynaptically, the A2AR antagonist tive intestinal peptide pituitary adenylate cyclase-activat-

SCH58261 in combination with the D1R antagonist ing polypeptide have been shown to inhibit mitochon-

SCH23390 has been shown to play a potentially neuro- drial apoptotic pathways, and protect neurons against

protective role in HD by decreasing glutamate release or oxidative stress-induced apoptosis and inflammation, pro-

enhancing glutamate uptake [144,145]. In contrast, viding an alternate avenue for therapeutic intervention in

SCH58261 and SCH23390 have been shown to promote neurodegenerative diseases [147,148]. Collectively, the

neurotoxicity when acting on postsynaptic A2ARs [145]. evidence indicates several viable avenues for therapeutic

In addition, the A2AR agonist CGS21680 has been shown intervention in neurodegenerative diseases.

to be neuroprotective by reducing NMDA currents in

stiatal medium spiny neurons [145] and to delay the onset Conflict of interest statement

of motor deterioration in R6/2 mice [146]. Thus, A2AR Nothing declared.

agonists and antagonists appear to provide some protec-

tion in animal models of HD. Although the A2AR is clearly References and recommended reading

involved in the pathophysiology of HD, further investi- Papers of particular interest, published within the period of review,

have been highlighted as:

gation into whether activation or inhibition of the A2AR is



warranted to establish the most advantageous avenue for of special interest

 of outstanding interest

therapeutic benefit in HD.

1. Przedborski S, Vila M, Jackson-Lewis V: Neurodegeneration:

Conclusion what is it and where are we? J Clin Invest 2003, 111:3-10.

Current therapeutic strategies provide temporary symp-

2. Arlt S: Non-Alzheimer’s disease-related memory impairment

tomatic relief but do not target the underlying pathobiol- and dementia. Dialogues Clin Neurosci 2013, 15:465-473.

ogy of the neurodegenerative diseases discussed here and 3. Dickerson BC, Bakkour A, Salat DH, Feczko E, Pacheco J,

Greve DN, Grodstein F, Wright CI, Blacker D, Rosas HD et al.: The

thus do not affect disease progression. In the current

cortical signature of Alzheimer’s disease: regionally specific

work, we summarize studies on several GPCRs that are

cortical thinning relates to symptom severity in very mild to

expressed in degenerative brain regions involved in AD, mild AD dementia and is detectable in asymptomatic amyloid-

positive individuals. Cereb Cortex 2009, 19:497-510.

VaD, FTD, PD, and HD and present the current evi-

4. Lopez OL: The growing burden of Alzheimer’s disease. Am J

dence, which supports therapeutic intervention strategies

Manag Care 2011, 17(Suppl. 13):S339-S345.

focused on functional modulation of specific GPCRs.

5. Serrano-Pozo A, Frosch MP, Masliah E, Hyman BT:

Neuropathological alterations in Alzheimer disease. Cold

An increasing number of GPCRs are being identified, Spring Harb Perspect Med 2011, 1:a006189.

which are involved in modulation of the neuropathologi-

6. Sachdev P, Kalaria R, O’Brien J, Skoog I, Alladi S, Black SE,

cal changes observed in neurodegenerative diseases. Blacker D, Blazer DG, Chen C, Chui H et al.: Diagnostic criteria

www.sciencedirect.com Current Opinion in Pharmacology 2017, 32:96–110

106 Neurosciences

for vascular cognitive disorders: a VASCOG statement. 25. Dong XX, Wang Y, Qin ZH: Molecular mechanisms of

Alzheimer Dis Assoc Disord 2014, 28:206-218. excitotoxicity and their relevance to pathogenesis of

neurodegenerative diseases. Acta Pharmacol Sin 2009,

7. Kalaria RN: Neuropathological diagnosis of vascular cognitive 30:379-387.

 impairment and vascular dementia with implications for

Alzheimer’s disease. Acta Neuropathol 2016, 131:659-685. 26. Porsteinsson AP, Cosman KM: Memantine in the treatment of

This review highlights the cerebrovascular pathologies of VaD and illus- Alzheimer’s disease. Aging Health 2006, 2:891-904.

trates which brain regions are affected by the various pathologies.

27. Danysz W, Parsons CG, Mobius HJ, Stoffler A, Quack G:

8. Yassi N, Desmond PM, Masters CL: Magnetic resonance Neuroprotective and symptomatological action of memantine

 imaging of vascular contributions to cognitive impairment and relevant for Alzheimer’s disease—a unified glutamatergic

dementia. J Mol Neurosci 2016, 60:349-353. hypothesis on the mechanism of action. Neurotox Res 2000,

This review highlights the different cerebrovascular pathologies in VaD 2:85-97.

and highlights the common brain regions that are affected in each

pathology. 28. Reisberg B, Doody R, Stoffler A, Schmitt F, Ferris S, Mobius HJ,

Memantine Study Group: Memantine in moderate-to-severe

9. Warren JD, Rohrer JD, Rossor MN: Frontotemporal dementia. Alzheimer’s disease. N Engl J Med 2003, 348:1333-1341.

BMJ 2013, 347:f4827.

29. Willard SS, Koochekpour S: Glutamate, glutamate receptors,

10. Jicha GA, Nelson PT: Management of frontotemporal dementia: and downstream signaling pathways. Int J Biol Sci 2013,

targeting symptom management in such a heterogeneous 9:948-959.

disease requires a wide range of therapeutic options.

30. Hamilton A, Esseltine JL, DeVries RA, Cregan SP, Ferguson SS:

Neurodegener Dis Manag 2011, 1:141-156.

Metabotropic glutamate receptor 5 knockout reduces

11. Gabryelewicz T, Masellis M, Berdynski M, Bilbao JM, Rogaeva E, cognitive impairment and pathogenesis in a mouse model of

St George-Hyslop P, Barczak A, Czyzewski K, Barcikowska M, Alzheimer’s disease. Mol Brain 2014, 7:40.

Wszolek Z et al.: Intra-familial clinical heterogeneity due to

31. Um JW, Kaufman AC, Kostylev M, Heiss JK, Stagi M, Takahashi H,

FTLD-U with TDP-43 proteinopathy caused by a novel deletion

Kerrisk ME, Vortmeyer A, Wisniewski T, Koleske AJ et al.:

in progranulin gene (PGRN). J Alzheimers Dis 2010,

22:1123-1133. Metabotropic glutamate receptor 5 is a coreceptor for

Alzheimer abeta oligomer bound to cellular prion protein.

12. Bertram L, Tanzi RE: The genetic epidemiology of Neuron 2013, 79:887-902.

neurodegenerative disease. J Clin Invest 2005, 115:1449-1457.

32. Hamilton A, Vasefi M, Vander Tuin C, McQuaid RJ, Anisman H,

13. Kalia LV, Lang AE: Parkinson’s disease. Lancet 2015, Ferguson SS: Chronic pharmacological mGluR5 inhibition

386:896-912. prevents cognitive impairment and reduces pathogenesis in

an Alzheimer disease mouse model. Cell Rep 2016,

14. Ross CA, Aylward EH, Wild EJ, Langbehn DR, Long JD, 15:1859-1865.

Warner JH, Scahill RI, Leavitt BR, Stout JC, Paulsen JS et al.:

Huntington disease: natural history, biomarkers and 33. Lo AC, De Maeyer JH, Vermaercke B, Callaerts-Vegh Z,

prospects for therapeutics. Nat Rev Neurol 2014, 10:204-216. Schuurkes JA, D’Hooge R: SSP-002392, a new 5-HT4 receptor

agonist, dose-dependently reverses scopolamine-induced

15. Andrew SE, Goldberg YP, Kremer B, Telenius H, Theilmann J, learning and memory impairments in C57Bl/6 mice.

Adam S, Starr E, Squitieri F, Lin B, Kalchman MA et al.: The Neuropharmacology 2014, 85:178-189.

relationship between trinucleotide (CAG) repeat length and

34. Zhang G, Asgeirsdottir HN, Cohen SJ, Munchow AH, Barrera MP,

clinical features of Huntington’s disease. Nat Genet 1993,

4:398-403. Stackman RW Jr: Stimulation of serotonin 2A receptors

facilitates consolidation and extinction of fear memory in

16. Gutekunst CA, Li SH, Yi H, Mulroy JS, Kuemmerle S, Jones R, C57BL/6J mice. Neuropharmacology 2013, 64:403-413.

Rye D, Ferrante RJ, Hersch SM, Li XJ: Nuclear and neuropil

35. Barthet G, Framery B, Gaven F, Pellissier L, Reiter E, Claeysen S,

aggregates in Huntington’s disease: relationship to

Bockaert J, Dumuis A: 5-Hydroxytryptamine 4 receptor

neuropathology. J Neurosci 1999, 19:2522-2534.

activation of the extracellular signal-regulated kinase pathway

17. Guerram M, Zhang LY, Jiang ZZ: G-protein coupled receptors as depends on Src activation but not on G protein or

therapeutic targets for neurodegenerative and beta-arrestin signaling. Mol Biol Cell 2007, 18:1979-1991.

cerebrovascular diseases. Neurochem Int 2016, 101:1-14.

36. Gooz M, Gooz P, Luttrell LM, Raymond JR: 5-HT2A receptor

18. Heng BC, Aubel D, Fussenegger M: An overview of the diverse induces ERK phosphorylation and proliferation through

roles of G-protein coupled receptors (GPCRs) in the ADAM-17 tumor necrosis factor-alpha-converting enzyme

pathophysiology of various human diseases. Biotechnol Adv (TACE) activation and heparin-bound epidermal growth

2013, 31:1676-1694. factor-like growth factor (HB-EGF) shedding in mesangial

cells. J Biol Chem 2006, 281:21004-21012.

19. Pierce KL, Premont RT, Lefkowitz RJ: Seven-transmembrane

receptors. Nat Rev Mol Cell Biol 2002, 3:639-650. 37. Misane I, Ogren SO: Selective 5-HT1A antagonists WAY

100635 and NAD-299 attenuate the impairment of passive

20. Vassilatis DK, Hohmann JG, Zeng H, Li F, Ranchalis JE, avoidance caused by scopolamine in the rat.

Mortrud MT, Brown A, Rodriguez SS, Weller JR, Wright AC et al.: Neuropsychopharmacology 2003, 28:253-264.

The G protein-coupled receptor repertoires of human and

mouse. Proc Natl Acad Sci U S A 2003, 100:4903-4908. 38. Yamazaki M, Okabe M, Yamamoto N, Yarimizu J, Harada K: Novel

5-HT5A receptor antagonists ameliorate scopolamine-

21. Millar RP, Newton CL: The year in G protein-coupled receptor induced working memory deficit in mice and reference

research. Mol Endocrinol 2010, 24:261-274. memory impairment in aged rats. J Pharmacol Sci 2015,

127:362-369.

22. Du AT, Schuff N, Amend D, Laakso MP, Hsu YY, Jagust WJ,

Yaffe K, Kramer JH, Reed B, Norman D et al.: Magnetic 39. Francken BJ, Jurzak M, Vanhauwe JF, Luyten WH, Leysen JE: The

resonance imaging of the entorhinal cortex and hippocampus human 5-ht5A receptor couples to Gi/Go proteins and inhibits

in mild cognitive impairment and Alzheimer’s disease. J Neurol adenylate cyclase in HEK 293 cells. Eur J Pharmacol 1998,

Neurosurg Psychiatry 2001, 71:441-447. 361:299-309.

23. Colovic MB, Krstic DZ, Lazarevic-Pasti TD, Bondzic AM, Vasic VM: 40. Gonzalez R, Chavez-Pascacio K, Meneses A: Role of 5-HT5A

Acetylcholinesterase inhibitors: pharmacology and receptors in the consolidation of memory. Behav Brain Res

toxicology. Curr Neuropharmacol 2013, 11:315-335. 2013, 252:246-251.

24. Selkoe DJ: The therapeutics of Alzheimer’s disease: where 41. Woods S, Clarke NN, Layfield R, Fone KC: 5-HT(6) receptor

we stand and where we are heading. Ann Neurol 2013, agonists and antagonists enhance learning and memory in a

74:328-336. conditioned emotion response paradigm by modulation of

Current Opinion in Pharmacology 2017, 32:96–110 www.sciencedirect.com

Role of GPCRs in neurodegenerative diseases Huang, Todd and Thathiah 107

cholinergic and glutamatergic mechanisms. Br J Pharmacol production by regulating gamma-secretase activity. EMBO J

2012, 167:436-449. 2015, 34:1674-1686.

This study demonstrates that CRF increases Ab production through

42. Ramirez MJ: 5-HT6 receptors and Alzheimer’s disease.

modulation of g-secrease function. The effect can be mediated by both

Alzheimer’s Res Ther 2013, 5:15.

CRF receptor-dependent and independent pathways.

43. Lu B, Nagappan G, Lu Y: BDNF and synaptic plasticity,

58. Rapp MA, Schnaider-Beeri M, Grossman HT, Sano M, Perl DP,

cognitive function, and dysfunction. Handb Exp Pharmacol

Purohit DP, Gorman JM, Haroutunian V: Increased hippocampal

2014, 220:223-250.

plaques and tangles in patients with Alzheimer disease with a

lifetime history of major depression. Arch Gen Psychiatry 2006,

44. Marcos B, Cabero M, Solas M, Aisa B, Ramirez MJ: Signalling

63:161-167.

pathways associated with 5-HT6 receptors: relevance for

cognitive effects. Int J Neuropsychopharmacol 2010,

59. Campbell SN, Zhang C, Roe AD, Lee N, Lao KU, Monte L,

13:775-784.

 Donohue MC, Rissman RA: Impact of CRFR1 ablation on

amyloid-beta production and accumulation in a mouse model

45. Albasanz JL, Perez S, Barrachina M, Ferrer I, Martin M:

of Alzheimer’s disease. J Alzheimer’s Dis 2015, 45:1175-1184.

Up-regulation of adenosine receptors in the frontal cortex in

This study uses a genetic approach to validate the CRF receptor type 1 in

Alzheimer’s disease. Brain Pathol 2008, 18:211-219.

an AD transgenic mouse model, demonstrating a reduction in amyloid

pathology in the absence of the CRF receptor type 1.

46. Borota D, Murray E, Keceli G, Chang A, Watabe JM, Ly M,

Toscano JP, Yassa MA: Post-study caffeine administration

60. Dong H, Wang S, Zeng Z, Li F, Montalvo-Ortiz J, Tucker C,

enhances memory consolidation in humans. Nat Neurosci

Akhtar S, Shi J, Meltzer HY, Rice KC et al.: Effects of

2014, 17:201-203.

corticotrophin-releasing factor receptor 1 antagonists on

47. Arendash GW, Schleif W, Rezai-Zadeh K, Jackson EK, amyloid-beta and behavior in Tg2576 mice.

Zacharia LC, Cracchiolo JR, Shippy D, Tan J: Caffeine protects Psychopharmacology (Berl) 2014, 231:4711-4722.

Alzheimer’s mice against cognitive impairment and reduces

61. Bale TL, Vale WW: Increased depression-like behaviors in

brain beta-amyloid production. Neuroscience 2006,

142:941-952. corticotropin-releasing factor receptor-2-deficient mice:

sexually dichotomous responses. J Neurosci 2003,

48. Dall’Igna OP, Fett P, Gomes MW, Souza DO, Cunha RA, Lara DR: 23:5295-5301.

Caffeine and adenosine A(2a) receptor antagonists prevent

62. Reul JM, Holsboer F: On the role of corticotropin-releasing

beta-amyloid (25-35)-induced cognitive deficits in mice.

hormone receptors in anxiety and depression. Dialogues Clin

Exp Neurol 2007, 203:241-245.

Neurosci 2002, 4:31-46.

49. Orr AG, Hsiao EC, Wang MM, Ho K, Kim DH, Wang X, Guo W,

Localization of novel

 Kang J, Yu GQ, Adame A et al.: Astrocytic 63. Chalmers DT, Lovenberg TW, De Souza EB:

corticotropin-releasing factor receptor (CRF2) mRNA

A2A and Gs-coupled signaling regulate memory. Nat Neurosci

2015, 18:423-434. expression to specific subcortical nuclei in rat brain:

comparison with CRF1 receptor mRNA expression

This is the first study to demonstrate the involvement of an astrocytic . J Neurosci

GPCR in the modulation of cognitive function. The study also shows 1995, 15:6340-6350.

increased level of astrocytic A2A receptors in the human AD brain and in

64. Snowden JS, Neary D, Mann DM: Frontotemporal dementia.

an AD transgenic mouse model.

Br J Psychiatry 2002, 180:140-143.

50. Santschi LA, Zhang XL, Stanton PK: Activation of receptors

65. Neary D, Snowden JS, Gustafson L, Passant U, Stuss D, Black S,

negatively coupled to adenylate cyclase is required for

Freedman M, Kertesz A, Robert PH, Albert M et al.:

induction of long-term synaptic depression at Schaffer

Frontotemporal lobar degeneration: a consensus on clinical

collateral-CA1 synapses. J Neurobiol 2006, 66:205-219.

diagnostic criteria. Neurology 1998, 51:1546-1554.

51. Thathiah A, Horre K, Snellinx A, Vandewyer E, Huang Y,

66. Jesso S, Morlog D, Ross S, Pell MD, Pasternak SH, Mitchell DG,

Ciesielska M, De Kloe G, Munck S, De Strooper B: Beta-arrestin

Kertesz A, Finger EC: The effects of oxytocin on social

2 regulates Abeta generation and gamma-secretase activity in

cognition and behaviour in frontotemporal dementia. Brain

Alzheimer’s disease. Nat Med 2013, 19:43-49.

2011, 134:2493-2501.

52. Thathiah A, Spittaels K, Hoffmann M, Staes M, Cohen A, Horre K,

Vanbrabant M, Coun F, Baekelandt V, Delacourte A et al.: The 67. Donaldson ZR, Young LJ: Oxytocin, vasopressin, and the

orphan G protein-coupled receptor 3 modulates amyloid-beta neurogenetics of sociality. Science 2008, 322:900-904.

peptide generation in neurons. Science 2009, 323:946-951.

68. Baumgartner T, Heinrichs M, Vonlanthen A, Fischbacher U, Fehr E:

53. Huang Y, Skwarek-Maruszewska A, Horre K, Vandewyer E, Oxytocin shapes the neural circuitry of trust and trust

 Wolfs L, Snellinx A, Saito T, Radaelli E, Corthout N, Colombelli J adaptation in humans. Neuron 2008, 58:639-650.

et al.: Loss of GPR3 reduces the amyloid plaque burden and

69. Sobota R, Mihara T, Forrest A, Featherstone RE, Siegel SJ:

improves memory in Alzheimer’s disease mouse models. Sci

Oxytocin reduces amygdala activity, increases social

Transl Med 2015, 7:309ra164.

interactions, and reduces anxiety-like behavior irrespective of

This study extensively scrutinizes the validity of GPR3 as a therapeutic

NMDAR antagonism. Behav Neurosci 2015, 129:389-398.

target for AD in various AD mouse models including two AD knockin

models. This study also shows increased level of GPR3 in two cohorts of

70. Theodosis DT, Koksma JJ, Trailin A, Langle SL, Piet R, Lodder JC,

human AD brain samples.

Timmerman J, Mansvelder H, Poulain DA, Oliet SH et al.: Oxytocin

54. Zhao J, Deng Y, Jiang Z, Qing H: G protein-coupled receptors and estrogen promote rapid formation of functional GABA

(GPCRs) in Alzheimer’s disease: a focus on BACE1 related synapses in the adult supraoptic nucleus. Mol Cell Neurosci

GPCRs. Front Aging Neurosci 2016, 8:58. 2006, 31:785-794.

55. Thathiah A, De Strooper B: The role of G protein-coupled 71. Finger EC, MacKinley J, Blair M, Oliver LD, Jesso S, Tartaglia MC,

receptors in the pathology of Alzheimer’s disease. Nat Rev Borrie M, Wells J, Dziobek I, Pasternak S et al.: Oxytocin for

Neurosci 2011, 12:73-87. frontotemporal dementia: a randomized dose-finding study of

safety and tolerability. Neurology 2015, 84:174-181.

56. Ishitobi Y, Nakayama S, Yamaguchi K, Kanehisa M, Higuma H,

Maruyama Y, Ninomiya T, Okamoto S, Tanaka Y, Tsuru J et al.: 72. Franceschi M, Anchisi D, Pelati O, Zuffi M, Matarrese M,

Association of CRHR1 and CRHR2 with major depressive Moresco RM, Fazio F, Perani D: Glucose metabolism and

disorder and panic disorder in a Japanese population. Am J serotonin receptors in the frontotemporal lobe degeneration.

Med Genet B Neuropsychiatr Genet 2012, 159b:429-436. Ann Neurol 2005, 57:216-225.

57. Park HJ, Ran Y, Jung JI, Holmes O, Price AR, Smithson L, 73. Procter AW, Qurne M, Francis PT: Neurochemical features of

 Ceballos-Diaz C, Han C, Wolfe MS, Daaka Y et al.: The stress frontotemporal dementia. Dement Geriatr Cogn Disord 1999,

response neuropeptide CRF increases amyloid-beta 10(Suppl. 1):80-84.

www.sciencedirect.com Current Opinion in Pharmacology 2017, 32:96–110

108 Neurosciences

74. Bowen DM, Procter AW, Mann DM, Snowden JS, Esiri MM, treating apathy in Alzheimer’s disease. Eur Psychiatry 2014,

Neary D, Francis PT: Imbalance of a serotonergic system in 29 1-1.

frontotemporal dementia: implication for pharmacotherapy.

Psychopharmacology (Berl) 2008, 196:603-610. 91. Avila A, Cardona X, Martin-Baranera M, Leon L, Caballol N,

Millet P, Bello J: Agomelatine for depression in Parkinson

75. Pompeiano M, Palacios JM, Mengod G: Distribution and cellular disease: additional effect on sleep and motor dysfunction.

localization of mRNA coding for 5-HT1A receptor in the rat J Clin Psychopharmacol 2015, 35:719-723.

brain: correlation with receptor binding. J Neurosci 1992,

12:440-453. 92. Roman GC, Erkinjuntti T, Wallin A, Pantoni L, Chui HC: Subcortical

ischaemic vascular dementia. Lancet Neurol 2002, 1:426-436.

76. King MV, Marsden CA, Fone KC: A role for the 5-HT(1A), 5-HT4

and 5-HT6 receptors in learning and memory. Trends 93. Boddeke EW, Enz A, Shapiro G: SDZ ENS 163, a selective

Pharmacol Sci 2008, 29:482-492. muscarinic M1 receptor agonist, facilitates the induction of

long-term potentiation in rat hippocampal slices. Eur J

77. Alagarsamy S, Marino MJ, Rouse ST, Gereau RWt, Heinemann SF, Pharmacol 1992, 222:21-25.

Conn PJ: Activation of NMDA receptors reverses

desensitization of mGluR5 in native and recombinant systems. 94. Tanaka K, Ogawa N, Asanuma M, Kondo Y, Nomura M:

Nat Neurosci 1999, 2:234-240. Relationship between cholinergic dysfunction and

discrimination learning disabilities in Wistar rats following

78. Alagarsamy S, Rouse ST, Junge C, Hubert GW, Gutman D, chronic cerebral hypoperfusion. Brain Res 1996, 729:55-65.

Smith Y, Conn PJ: NMDA-induced phosphorylation and

regulation of mGluR5. Pharmacol Biochem Behav 2002, 95. Kondo Y, Ogawa N, Asanuma M, Matsuura K, Nishibayashi K,

73:299-306. Iwata E: Preventive effects of bifemelane hydrochloride on

decreased levels of muscarinic acetylcholine receptor and its

79. Leuzy A, Zimmer ER, Dubois J, Pruessner J, Cooperman C, mRNA in a rat model of chronic cerebral hypoperfusion.

 Soucy JP, Kostikov A, Schirmaccher E, Desautels R, Gauthier S Neurosci Res 1996, 24:409-414.

et al.: In vivo characterization of metabotropic glutamate

96. Huang YY, Kandel ER: Age-related enhancement of a protein

receptor type 5 abnormalities in behavioral variant FTD. Brain

synthesis-dependent late phase of LTP induced by low

Struct Funct 2016, 221:1387-1402.

frequency paired-pulse stimulation in hippocampus. Learn

This study is the first to show a decrease in mGluR5 availability in bvFTD

Mem 2006, 13:298-306.

patients, which may lead to NMDAR hypofunction and neurotoxicity.

97. Rossato JI, Bevilaqua LR, Izquierdo I, Medina JH, Cammarota M:

80. Homayoun H, Moghaddam B: Bursting of prefrontal cortex

Dopamine controls persistence of long-term memory storage.

neurons in awake rats is regulated by metabotropic glutamate

Science 2009, 325:1017-1020.

5 (mGlu5) receptors: rate-dependent influence and interaction

with NMDA receptors. Cereb Cortex 2006, 16:93-105.

98. Wan P, Wang S, Zhang Y, Lv J, Jin QH: Involvement of dopamine

 D1 receptors of the hippocampal dentate gyrus in spatial

81. Kinney GG, O’Brien JA, Lemaire W, Burno M, Bickel DJ,

learning and memory deficits in a rat model of vascular

Clements MK, Chen TB, Wisnoski DD, Lindsley CW, Tiller PR et al.:

dementia. Pharmazie 2014, 69:709-710.

A novel selective positive allosteric modulator of

This study provides evidence that a D1R agonist can attenuate the

metabotropic glutamate receptor subtype 5 has in vivo activity

cognitive impairments observed in a CCH rat model of VD.

and antipsychotic-like effects in rat behavioral models. J

Pharmacol Exp Ther 2005, 313:199-206.

99. Shahidi S, Komaki A, Mahmoodi M, Lashgari R: The role of

GABAergic transmission in the dentate gyrus on acquisition,

82. Hurd YL, Suzuki M, Sedvall GC: D1 and D2

consolidation and retrieval of an inhibitory avoidance learning

mRNA expression in whole hemisphere sections of the human

and memory task in the rat. Brain Res 2008, 1204:87-93.

brain. J Chem Neuroanat 2001, 22:127-137.

100. Li CJ, Lu Y, Zhou M, Zong XG, Li C, Xu XL, Guo LJ, Lu Q:

83. Seeman P: Atypical antipsychotics: mechanism of action.

Activation of GABAB receptors ameliorates cognitive

Can J Psychiatry 2002, 47:27-38.

impairment via restoring the balance of HCN1/HCN2 surface

84. Kapur S, Seeman P: Does fast dissociation from the dopamine expression in the hippocampal CA1 area in rats with chronic

d(2) receptor explain the action of atypical antipsychotics? A cerebral hypoperfusion. Mol Neurobiol 2014, 50:704-720.

new hypothesis. Am J Psychiatry 2001, 158:360-369.

101. Li G, Lv J, Wang J, Wan P, Li Y, Jiang H, Jin Q: GABAB receptors



85. Antonini A, Leenders KL, Spiegel R, Meier D, Vontobel P, Weigell- in the hippocampal dentate gyrus are involved in spatial

Weber M, Sanchez-Pernaute R, de Yebenez JG, Boesiger P, learning and memory impairment in a rat model of vascular

Weindl A et al.: Striatal glucose metabolism and dopamine D2 dementia. Brain Res Bull 2016, 124:190-197.

receptor binding in asymptomatic gene carriers and patients This study determined that GABAB receptor activity is elevated in the

with Huntington’s disease. Brain 1996, 119(Pt. 6):2085-2095. hippocampus of a VaD rat model. In conjunction with the contradictory

findings in reference 100, this study highlights the importance of studying

86. Kanazawa I, Kwak S, Sasaki H, Muramoto O, Mizutani T, Hori A, hippocampal GABABRs in VaD models.

Nukina N: Studies on neurotransmitter markers of the basal

ganglia in Pick’s disease, with special reference to dopamine 102. Chumakov I, Nabirotchkin S, Cholet N, Milet A, Boucard A,



reduction. J Neurol Sci 1988, 83:63-74. Toulorge D, Pereira Y, Graudens E, Traore S, Foucquier J et al.:

Combining two repurposed drugs as a promising approach for

87. Sperfeld AD, Collatz MB, Baier H, Palmbach M, Storch A, Alzheimer’s disease therapy. Sci Rep 2015, 5:7608.

Schwarz J, Tatsch K, Reske S, Joosse M, Heutink P et al.: This study demonstrates that a combination of two FDA-approved drugs

FTDP-17: an early-onset phenotype with parkinsonism and targeting excitatory and inhibitory systems alleviate the cognitive deficits

epileptic seizures caused by a novel mutation. Ann Neurol in AD mouse models.

1999, 46:708-715.

103. Bennys K, Haddad R, Gres CS, Schmitt P, Touchon J: Cognitive

88. Pagonabarraga J, Kulisevsky J, Strafella AP, Krack P: Apathy in event-related potentials used as biomarkers in Pleodial-I

Parkinson’s disease: clinical features, neural substrates, study: first evidence of a neurophysiological effect of PXT864

diagnosis, and treatment. Lancet Neurol 2015, 14:518-531. in mild Alzheimer’s disease patients. Alzheimer’s Dement J

Alzheimer’s Assoc 2016, 12:P1061.

89. Callegari I, Mattei C, Benassi F, Krueger F, Grafman J, Yaldizli O,

 Sassos D, Massucco D, Scialo C, Nobili F et al.: Agomelatine 104. Pompeiano M, Palacios JM, Mengod G: Distribution of the

improves apathy in frontotemporal dementia. Neurodegener serotonin 5-HT2 receptor family mRNAs: comparison between

Dis 2016, 16:352-356. 5-HT2A and 5-HT2C receptors. Brain Res Mol Brain Res 1994,

This study introduces agomelatine, a weak 5-HT2C receptor antagonist, 23:163-178.

as a potential treatment for apathy in FTD patients.

105. Glikmann-Johnston Y, Saling MM, Reutens DC, Stout JC:

90. Karaiskos DPD, Katirtzoglou E, Siarkos K, Tzavellas E, Hippocampal 5-HT1A receptor and spatial learning and

Papadimitrious GN, Politis A: EPA-1197-agomelatine for memory. Front Pharmacol 2015, 6:289.

Current Opinion in Pharmacology 2017, 32:96–110 www.sciencedirect.com

Role of GPCRs in neurodegenerative diseases Huang, Todd and Thathiah 109

106. Elliott MS, Ballard CG, Kalaria RN, Perry R, Hortobagyi T, and restless legs syndrome. Drugs Today (Barc) 2010, 46:483-

Francis PT: Increased binding to 5-HT1A and 5-HT2A receptors 505.

is associated with large vessel infarction and relative

123. Brooks DJ: Dopamine agonists: their role in the treatment of

preservation of cognition. Brain 2009, 132:1858-1865.

Parkinson’s disease. J Neurol Neurosurg Psychiatry 2000,

107. Roman GC, Wilkinson DG, Doody RS, Black SE, Salloway SP, 68:685-689.

Schindler RJ: Donepezil in vascular dementia: combined

124. Mori A, LeWitt P, Jenner P: The story of istradefylline—the first

analysis of two large-scale clinical trials. Dement Geriatr Cogn

 approved A2A antagonist for the treatment of Parkinson’s

Disord 2005, 20:338-344.

disease. In The Adenosinergic System: A Non-Dopaminergic

108. Baskys A, Hou AC: Vascular dementia: pharmacological Target in Parkinson’s Disease. Edited by Morelli M, Simola N,

treatment approaches and perspectives. Clin Interv Aging 2007, Wardas J. Springer International Publishing; 2015:273-289.

2:327-335. This work summarizes the development of istradefylline for the treatment

of Parkinson’s disease.

109. Sveinbjornsdottir S: The clinical symptoms of Parkinson’s

disease. J Neurochem 2016, 139(Suppl. 1):318-324. 125. Uchida S, Soshiroda K, Okita E, Kawai-Uchida M, Mori A,

Jenner P, Kanda T: The antagonist,

110. Aarsland D, Bronnick K, Williams-Gray C, Weintraub D, Marder K,

istradefylline enhances anti-parkinsonian activity induced by

Kulisevsky J, Burn D, Barone P, Pagonabarraga J, Allcock L et al.:

combined treatment with low doses of L-DOPA and dopamine

Mild cognitive impairment in Parkinson disease: a multicenter

agonists in MPTP-treated common marmosets. Eur J

pooled analysis. Neurology 2010, 75:1062-1069.

Pharmacol 2015, 766:25-30.

111. Wood K-L, Myall DJ, Livingston L, Melzer TR, Pitcher TL,

126. Lester DB, Rogers TD, Blaha CD: Acetylcholine-dopamine

 MacAskill MR, Geurtsen JG, Anderson TJ, Dalrymple-Alford JC:

interactions in the pathophysiology and treatment of CNS

Different PD-MCI criteria and risk of dementia in Parkinson’s

disorders. CNS Neurosci Ther 2010, 16:137-162.

disease: 4-year longitudinal study. npj Parkinson’s Dis 2016.

This study presents the criteria from a longitudinal study to predict the risk 127. Fink-Jensen A, Schmidt LS, Dencker D, Schulein C, Wess J,

to develop PDD from PD-MCI and requires at least two impairments Wortwein G, Woldbye DP: Antipsychotic-induced catalepsy is

within a single cognitive domain. attenuated in mice lacking the M4 muscarinic acetylcholine

receptor. Eur J Pharmacol 2011, 656:39-44.

112. Hely MA, Reid WG, Adena MA, Halliday GM, Morris JG: The

Sydney multicenter study of Parkinson’s disease: the 128. Svenningsson P, Westman E, Ballard C, Aarsland D: Cognitive

inevitability of dementia at 20 years. Mov Disord 2008, 23:837- impairment in patients with Parkinson’s disease: diagnosis,

844. biomarkers, and treatment. Lancet Neurol 2012, 11:697-707.

113. Mak E, Bergsland N, Dwyer MG, Zivadinov R, Kandiah N: 129. Marazziti D, Golini E, Mandillo S, Magrelli A, Witke W, Matteoni R,

Subcortical atrophy is associated with cognitive impairment in Tocchini-Valentini GP: Altered dopamine signaling and MPTP

mild Parkinson disease: a combined investigation of resistance in mice lacking the Parkinson’s disease-

volumetric changes, cortical thickness, and vertex-based associated GPR37/parkin-associated endothelin-like

shape analysis. AJNR Am J Neuroradiol 2014, 35:2257-2264. receptor. Proc Natl Acad Sci U S A 2004, 101:10189-10194.

114. Hanganu A, Bedetti C, Degroot C, Mejia-Constain B, 130. Wu CS, Chen H, Sun H, Zhu J, Jew CP, Wager-Miller J, Straiker A,

 Lafontaine AL, Soland V, Chouinard S, Bruneau MA, Mellah S, Spencer C, Bradshaw H, Mackie K et al.: GPR55, a G-protein

Belleville S et al.: Mild cognitive impairment is linked with faster coupled receptor for lysophosphatidylinositol, plays a role in

rate of cortical thinning in patients with Parkinson’s disease motor coordination. PLoS One 2013, 8:e60314.

longitudinally. Brain 2014, 137:1120-1129.

131. Gandia J, Morato X, Stagljar I, Fernandez-Duenas V, Ciruela F:

This study reveals the correlation between the reduced volume in the

Adenosine A2A receptor-mediated control of pilocarpine-

nucleus accumbens and amydala in PD with mild cognitive impairment

(MCI). induced tremulous jaw movements is Parkinson’s disease-

associated GPR37 receptor-dependent. Behav Brain Res 2015,

115. Beaulieu JM, Gainetdinov RR: The physiology, signaling, and 288:103-106.

pharmacology of dopamine receptors. Pharmacol Rev 2011,

63:182-217. 132. Beal MF, Ferrante RJ, Swartz KJ, Kowall NW: Chronic quinolinic

acid lesions in rats closely resemble Huntington’s disease.

116. Hisahara S, Shimohama S: Dopamine receptors and J Neurosci 1991, 11:1649-1659.

Parkinson’s disease. Int J Med Chem 2011, 2011:403039.

133. Trepanier C, Lei G, Xie YF, MacDonald JF: Group II metabotropic

117. Pezze MA, Dalley JW, Robbins TW: Differential roles of glutamate receptors modify N-methyl-D-aspartate receptors

dopamine D1 and D2 receptors in the nucleus accumbens in via Src kinase. Sci Rep 2013, 3:926.

attentional performance on the five-choice serial reaction

134. Bruno V, Ksiazek I, Battaglia G, Lukic S, Leonhardt T, Sauer D,

time task. Neuropsychopharmacology 2007, 32:273-283.

Gasparini F, Kuhn R, Nicoletti F, Flor PJ: Selective blockade of

118. Ingallinesi M, Le Bouil L, Biguet NF, Thi AD, Mannoury la Cour C, metabotropic glutamate receptor subtype 5 is

 Millan MJ, Ravassard P, Mallet J, Meloni R: Local inactivation of neuroprotective. Neuropharmacology 2000, 39:2223-2230.

Gpr88 in the nucleus accumbens attenuates behavioral

135. Schiefer J, Sprunken A, Puls C, Luesse HG, Milkereit A, Milkereit E,

deficits elicited by the neonatal administration of

Johann V, Kosinski CM: The metabotropic glutamate receptor

phencyclidine in rats. Mol Psychiatry 2015, 20:951-958.

5 antagonist MPEP and the mGluR2 agonist LY379268 modify

This study demonstrates the specific role of the dopamine D2 receptor in

disease progression in a transgenic mouse model of

the rat nucleus accumbens by reducing receptor expression using

microRNAs. Huntington’s disease. Brain Res 2004, 1019:246-254.

136. Yao Y, Cui X, Al-Ramahi I, Sun X, Li B, Hou J, Difiglia M, Palacino J,

119. Alegret M, Junque C, Pueyo R, Valldeoriola F, Vendrell P, Tolosa E,

 Wu ZY, Ma L et al.: A striatal-enriched intronic GPCR modulates

Mercader JM: MRI atrophy parameters related to cognitive and

huntingtin levels and toxicity. Elife 2015, 4.

motor impairment in Parkinson’s disease. Neurologia 2001,

16:63-69. This study demonstrates that GPR52 stabilizes mutant huntingtin by

preventing degradation. Reduced levels of GPR52 suppress HD pheno-

120. Chase TN, Mouradian MM, Engber TM: Motor response types in both patient iPS-derived neurons and Drosophila HD models.

complications and the function of striatal efferent systems.

137. Chen JY, Wang EA, Cepeda C, Levine MS: Dopamine imbalance

Neurology 1993, 43:S23-S27.

in Huntington’s disease: a mechanism for the lack of

121. Bonuccelli U, Del Dotto P, Rascol O: Role of dopamine receptor behavioral flexibility. Front Neurosci 2013, 7:114.

agonists in the treatment of early Parkinson’s disease.

138. Charvin D, Vanhoutte P, Pages C, Borrelli E, Caboche J:

Parkinsonism Relat Disord 2009, 15(Suppl. 4):S44-S53.

Unraveling a role for dopamine in Huntington’s disease: the

122. Boroojerdi B, Wolff HM, Braun M, Scheller DK: Rotigotine dual role of reactive oxygen species and D2 receptor

transdermal patch for the treatment of Parkinson’s disease stimulation. Proc Natl Acad Sci U S A 2005, 102:12218-12223.

www.sciencedirect.com Current Opinion in Pharmacology 2017, 32:96–110

110 Neurosciences

139. Charvin D, Roze E, Perrin V, Deyts C, Betuing S, Pages C, 155. Ribeiro FM, Devries RA, Hamilton A, Guimaraes IM, Cregan SP,

Regulier E, Luthi-Carter R, Brouillet E, Deglon N et al.: Haloperidol  Pires RG, Ferguson SS: Metabotropic glutamate receptor

protects striatal neurons from dysfunction induced by 5 knockout promotes motor and biochemical alterations in a

mutated huntingtin in vivo. Neurobiol Dis 2008, 29:22-29. mouse model of Huntington’s disease. Hum Mol Genet 2014,

23:2030-2042.

140. Tang TS, Chen X, Liu J, Bezprozvanny I: Dopaminergic signaling

This study demonstrates that the metabotropic glutamate receptor 5 is

and striatal neurodegeneration in Huntington’s disease.

involved in locomotor activity in HD mice by genetic and pharmacological

J Neurosci 2007, 27:7899-7910. approaches.

141. Katona I, Freund TF: Endocannabinoid signaling as a synaptic

156. Zhu D, Li C, Swanson AM, Villalba RM, Guo J, Zhang Z, Matheny S,

circuit breaker in neurological disease. Nat Med 2008,

Murakami T, Stephenson JR, Daniel S et al.: BAI1 regulates

14:923-930.

spatial learning and synaptic plasticity in the hippocampus. J

Clin Invest 2015, 125:1497-1508.

142. Blazquez C, Chiarlone A, Sagredo O, Aguado T, Pazos MR,

Resel E, Palazuelos J, Julien B, Salazar M, Borner C et al.: Loss of

157. Marino MJ, Williams DL Jr, O’Brien JA, Valenti O, McDonald TP,

striatal type 1 cannabinoid receptors is a key pathogenic

Clements MK, Wang R, DiLella AG, Hess JF, Kinney GG et al.:

factor in Huntington’s disease. Brain 2011, 134:119-136.

Allosteric modulation of group III metabotropic glutamate

receptor 4: a potential approach to Parkinson’s disease

143. Scotter EL, Goodfellow CE, Graham ES, Dragunow M, Glass M:

treatment. Proc Natl Acad Sci U S A 2003, 100:13668-13673.

Neuroprotective potential of CB1 receptor agonists in an in

vitro model of Huntington’s disease. Br J Pharmacol 2010,

160:747-761. 158. Niswender CM, Johnson KA, Weaver CD, Jones CK, Xiang Z,

Luo Q, Rodriguez AL, Marlo JE, de Paulis T, Thompson AD et al.:

144. Gomes CV, Kaster MP, Tome AR, Agostinho PM, Cunha RA: Discovery, characterization, and antiparkinsonian effect of

Adenosine receptors and brain diseases: neuroprotection and novel positive allosteric modulators of metabotropic

neurodegeneration. Biochim Biophys Acta 1808, 2011:1380- glutamate receptor 4. Mol Pharmacol 2008, 74:1345-1358.

1399.

159. Tang SS, Ji MJ, Chen L, Hu M, Long Y, Li YQ, Miao MX, Li JC, Li N,

145. Tebano MT, Pintor A, Frank C, Domenici MR, Martire A, Pepponi R, Ji H et al.: Protective effect of pranlukast on Abeta(1)(À)(4)(2)-

Potenza RL, Grieco R, Popoli P: Adenosine A2A receptor induced cognitive deficits associated with downregulation of

blockade differentially influences excitotoxic mechanisms at cysteinyl leukotriene receptor 1. Int J Neuropsychopharmacol

pre- and postsynaptic sites in the rat striatum. J Neurosci Res 2014, 17:581-592.

2004, 77:100-107.

160. Teng L, Zhao J, Wang F, Ma L, Pei G: A GPCR/secretase

146. Chou SY, Lee YC, Chen HM, Chiang MC, Lai HL, Chang HH, complex regulates beta- and gamma-secretase specificity for

Wu YC, Sun CN, Chien CL, Lin YS et al.: CGS21680 attenuates Abeta production and contributes to AD pathogenesis.

symptoms of Huntington’s disease in a transgenic mouse Cell Res 2010, 20:138-153.

model. J Neurochem 2005, 93:310-320.

161. Heaney CF, Kinney JW: Role of GABA(B) receptors in learning

147. Lee EH, Seo SR: Neuroprotective roles of pituitary adenylate

and memory and neurological disorders. Neurosci Biobehav

cyclase-activating polypeptide in neurodegenerative

Rev 2016, 63:1-28.

diseases. BMB Rep 2014, 47:369-375.

162. Faivre E, Gault VA, Thorens B, Holscher C: Glucose-dependent

148. White CM, Ji S, Cai H, Maudsley S, Martin B: Therapeutic

insulinotropic polypeptide receptor knockout mice are

potential of vasoactive intestinal peptide and its receptors in

impaired in learning, synaptic plasticity, and neurogenesis.

neurological disorders. CNS Neurol Disord Drug Targets 2010,

J Neurophysiol 2011, 105:1574-1580.

9:661-666.

163. Kubota T, Matsumoto H, Kirino Y: Ameliorative effect of

149. Price DL, Bonhaus DW, McFarland K: Pimavanserin, a 5-HT2A

membrane-associated estrogen receptor G protein coupled

receptor inverse agonist, reverses psychosis-like behaviors in

receptor 30 activation on object recognition memory in mouse

a rodent model of Alzheimer’s disease. Behav Pharmacol 2012,

models of Alzheimer’s disease. J Pharmacol Sci 2016, 23:426-433.

131:219-222.

150. Strange PG: Antipsychotic drugs: importance of dopamine

164. Yi T, Weng J, Siwko S, Luo J, Li D, Liu M: LGR4/GPR48

receptors for mechanisms of therapeutic actions and side

inactivation leads to aniridia-genitourinary anomalies-

effects. Pharmacol Rev 2001, 53:119-133.

mental retardation syndrome defects. J Biol Chem 2014,

151. Mievis S, Blum D, Ledent C: A2A receptor knockout worsens 289:8767-8780.

survival and motor behaviour in a transgenic mouse model of

165. Caccamo A, Oddo S, Billings LM, Green KN, Martinez-Coria H,

Huntington’s disease. Neurobiol Dis 2011, 41:570-576.

Fisher A, LaFerla FM: M1 receptors play a central role in

152. Popoli P, Blum D, Martire A, Ledent C, Ceruti S, Abbracchio MP: modulating AD-like pathology in transgenic mice.

Functions, dysfunctions and possible therapeutic relevance of Neuron 2006, 49:671-682.

adenosine A2A receptors in Huntington’s disease. Prog

Neurobiol 2007, 81:331-348. 166. Poulin B, Butcher A, McWilliams P, Bourgognon JM, Pawlak R,

Kong KC, Bottrill A, Mistry S, Wess J, Rosethorne EM et al.: The

153. Bannon NM, Zhang P, Ilin V, Chistiakova M, Volgushev M: M3-muscarinic receptor regulates learning and memory in a

Modulation of synaptic transmission by adenosine in layer 2/3 receptor phosphorylation/arrestin-dependent manner.

of the rat visual cortex in vitro. Neuroscience 2014, 260:171-184. Proc Natl Acad Sci U S A 2010, 107:9440-9445.

154. Pancani T, Foster DJ, Moehle MS, Bichell TJ, Bradley E,

167. Asle-Rousta M, Kolahdooz Z, Oryan S, Ahmadiani A, Dargahi L:

Bridges TM, Klar R, Poslusney M, Rook JM, Daniels JS et al.:

FTY720 (fingolimod) attenuates beta-amyloid peptide

Allosteric activation of M4 muscarinic receptors improve

(Abeta42)-induced impairment of spatial learning and memory

behavioral and physiological alterations in early symptomatic

in rats. J Mol Neurosci 2013, 50:524-532.

YAC128 mice. Proc Natl Acad Sci U S A 2015, 112:14078-14083.

Current Opinion in Pharmacology 2017, 32:96–110 www.sciencedirect.com