Next Article in Journal
Study on Demethoxycurcumin as a Promising Approach to Reverse Methicillin-Resistance of Staphylococcus aureus
Next Article in Special Issue
Deep Learning with Neuroimaging and Genomics in Alzheimer’s Disease
Previous Article in Journal
Application of the Antibody-Inducing Activity of Glycosphingolipids to Human Diseases
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Polymorphisms of Dopamine Receptor Genes and Parkinson’s Disease: Clinical Relevance and Future Perspectives

1
PhD Program in Clinical and Experimental Medicine and Medical Humanities, University of Insubria, 21100 Varese, Italy
2
Movement Disorders Centre, Neurology Unit, Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy
3
Centre of Research in Medical Pharmacology, University of Insubria, 21100 Varese, Italy
4
PhD Program in Medical Sciences and Biotechnology, University of Piemonte Orientale, 28100 Novara, Italy
5
Center of Research in Neuroscience, University of Insubria, 21100 Varese, Italy
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2021, 22(7), 3781; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22073781
Submission received: 1 March 2021 / Revised: 26 March 2021 / Accepted: 1 April 2021 / Published: 6 April 2021
(This article belongs to the Special Issue Genomics of Brain Disorders 3.0)

Abstract

:
Parkinson’s disease (PD) is a neurodegenerative disease caused by loss of dopaminergic neurons in the midbrain. PD is clinically characterized by a variety of motor and nonmotor symptoms, and treatment relies on dopaminergic replacement. Beyond a common pathological hallmark, PD patients may present differences in both clinical progression and response to drug therapy that are partly affected by genetic factors. Despite extensive knowledge on genetic variability of dopaminergic receptors (DR), few studies have addressed their relevance as possible influencers of clinical heterogeneity in PD patients. In this review, we summarized available evidence regarding the role of genetic polymorphisms in DR as possible determinants of PD development, progression and treatment response. Moreover, we examined the role of DR in the modulation of peripheral immunity, in light of the emerging role of the peripheral immune system in PD pathophysiology. A better understanding of all these aspects represents an important step towards the development of precise and personalized disease-modifying therapies for PD.

1. Introduction

Parkinson’s disease (PD) is a common neurodegenerative disease, with an overall prevalence of 0.3%, which tends to increase with age [1]. PD is clinically characterized by a wide array of motor and nonmotor manifestations that have a great impact on patients’ quality of life with both social and economic load [2].
In the initial disease stages, when diagnosis is usually established, motor symptoms include slowness of movement (bradykinesia), rest tremor and muscle rigidity. At this stage, patients may also complain of hyposmia, constipation, fatigue, sleep disturbances, mood deflection and impulse control disorder [3] As the disease progresses, motor complications may appear, consisting in wearing-off of the benefit provided by medication or even failure or delay of the medication effect. Involuntary choreiform movements appear when the blood levels of medication either peak or drop. The nonmotor features of this advanced disease stage may consist in visual hallucinations, cognitive decline and autonomic dysfunction (orthostatic hypotension, urinary dysfunction).
The pathological hallmark of PD is represented by the degeneration of the dopaminergic neurons in the pars compacta of the midbrain substantia nigra. Cell loss is accompanied by the accumulation of alpha-synuclein (α-syn) and it is currently believed that α-syn accumulation is correlated with PD progression [4]. Currently, the gold standard treatment for PD consists of dopamine (DA) replacement therapy (DRT) aiming at counterbalancing DA loss caused by nigrostriatal degeneration. Accordingly, the most used drugs for PD treatment are the DA precursor Levodopa, alone or together with MAO-B and COMT inhibitors, and DA-agonists. Unfortunately, as disease progresses, the benefits of symptomatic therapies tend to wear off [5] and to be counterbalanced by the onset of side effects and complications [6]
There are five types of DR, D1, D2, D3, D4 and D5, which are members of the G-protein coupled receptor family [7]. The dopaminergic receptor (DR) subtypes are divided into two families according to their pharmacological profile and second messenger coupling: the “D1-like”, including D1 and D5 which activate adenylate cyclase, and “D2-like” including types D2, D3 and D4 which inhibit adenylate cyclase [8]. The final effect of D1-like activation (D1 and D5) can be both excitation (via opening of sodium channels) and inhibition (via opening of potassium channels), while the ultimate effect of D2-like activation (D2, D3 and D4) is inhibition of target neuron [9].
D1 receptors are the most abundant DR in the human nervous system followed by D2 and other DR (D3, D4 and D5) whose levels are significantly lower [9].
Despite the key role of dopaminergic pathways in the pathogenesis as well as in the pharmacotherapy of PD, evidence on the role of genetic polymorphisms in DR and related pathways is still fragmentary. In this regard, several functional single nucleotide polymorphisms (SNPs, i.e., DNA sequence variations occurring when a single nucleotide in the genome differs between paired chromosomes), the most common type of polymorphisms in the human genome, have been identified in dopamine receptor genes (DR) [10,11]. Among these SNPs, some have been related with other neuropsychiatric conditions such as schizophrenia [12,13], attention deficit hyperactivity disorder [14,15], addictions [16] and even to clinical aspects of PD [17,18].
Furthermore, DR play a key role in the regulation of peripheral immunity [19,20,21] and recent findings suggest a role for the peripheral immune response in PD. For example, it has been shown that α-syn is recognized by T cells, thus suggesting a relationship between protein deposition, neuronal loss and immune response [4,22]. Since immune cells express DR, it is reasonable to suppose that SNPs in genes coding for these receptors could modulate functions of these cells. In this regard, it has been recently suggested that SNPs in DRs could influence immune cell functions in different ways [23,24]. Nonetheless, the relevance of such effects on PD development and progression, including response to therapy, has never been examined so far.
In the present review, we collected and critically appraised available evidence on the role of genetic polymorphisms in dopaminergic receptors with respect to: (i) risk of PD development, (ii) variability of PD phenotypes, (iii) response to pharmacological therapy and risk of side effect, (iv) immune function.

2. Pharmacogenetics of Dopaminergic Receptors

Several genetic polymorphisms have been described in DRs, and it had been suggested that DRs variability may be involved in both disease susceptibility and individual response to pharmacological treatments in PD.

2.1. Genetic Polymorphisms in Dopamine Receptor D1 Gene

The DR D1 gene (DRD1) contains two exons [25] and its alternative splicing results in the expression of two different transcripts. The genetic analysis of the DRD1 sequence has allowed the identification of various polymorphisms located within the gene whose functional consequences are still not fully understood. Among the most studied SNPs, rs686 contains a G-to-A substitution, with a frequency of the minor A allele of about 60% in Caucasian population. This SNP was associated with increased promoter activity and higher DRD1 expression. Another important SNP in DRD1 is rs5327, consisting in an A-to-C substitution (amino acid changing Thr37Pro), which was associated with reduction in total and surface receptor expression [26], while rs1799914, a G-to-A substitution, featured reduction in both mRNA expression levels and Bmax. This SNP was also associated with a reduced heterodimerisation of the D2 receptor [27].
Another important SNP in DRD1 is A-to-G substitution at position −48 in promoter region (rs4532); this SNP shown a frequency of 30% in Caucasian population, whereas for several other DRD1 SNPs, such as rs5330, rs5331, rs13306309, no biological or clinical functions have been described yet.

2.2. Genetic Polymorphisms in Dopamine Receptor D2 Gene

The DR D2 gene (DRD2) is located on chromosome 11q23 and contains six introns. Through the alternative splicing, two main variants are generated, named D2S and D2L [28].
More than 200 polymorphisms have been identified in DRD2, mostly in the introns and in downstream flanking region [29], which have been associated with addictions (alcohol, cocaine, nicotine and opioid), mood disorders, schizophrenia, movement disorders and drug response (reviewed in [30,31]).
rs1800497 (also known as TAQ1 A), the most studied DRD2 polymorphism, consists of a C-to-T substitution (amino acid changing Glu713Lys), with a frequency of the minor T allele of about 22% in Caucasian population. This variation was associated with a 40% reduction in D2 receptors expression in the striatum, without affecting receptor affinity [32,33,34,35].
A second important polymorphism in DRD2 is −141 C Ins/Del (rs1799732). This SNP is located in the promoter region of the DRD2 with allele frequency of about 9% of the Caucasian population [36] and is associated with a higher DR density in striatum [34].
Another frequent SNP in DRD2 is rs6277 which consists of a C-to-T substitution at positions 957 located in exon 7. The minor allele (T) has a frequency of about 50% in Caucasian population. The SNP was associated with decreased mRNA stability and translation, reduced DA-induced up-regulation of DR D2 membrane expression in vitro [37], and lower DR D2 expression in cortex and striatum of healthy subjects [38,39,40].
SNPs rs2283265 (consisting of a C-to-G substitution) and rs1076560 (consisting of a G-to-T substitution) alter mRNA splicing and transcription process in exon 6 leading to two isoforms of DRD2, which are D2 long and D2 short and were associated with low mRNA expression levels [31,41,42,43,44]. More specifically, these SNPs were associated with reduced DRD2 expression in prefrontal cortex and striatum as well as with altered activity of the striato-thalamic-prefrontal pathway in healthy subjects [31,45] and in schizophrenia [41]. Two other SNPs were associated with reductions in DRD2 gene activity, in particular rs1801028 (consisting of a C-to-G substitution, amino acid changing Ser311Cys) was associated with reduced DA affinity [46], and rs1079597 (consisting of a G-to-A substitution) with lower receptor binding [32,34]. On the contrary, SNP rs12364283 (consisting of a T-to-C substitution) was associated with increase in DRD2 receptor activity through an increase in mRNA expression levels [47].
Finally, rs2734849, a G-to-A transition, produces an amino-acid change (arginine to histidine) in C-terminal ankyrin repeat domain of ANKK1. Using the luciferase reporter assay, it has been demonstrated that this SNP alters expression level of NF-kB-regulated genes. Since DRD2 expression is regulated by transcription factor NF-kB, it has been postulated that rs2734849 may indirectly affect DRD2 density, although this hypothesis has not yet been confirmed in ad-hoc study [48].

2.3. Genetic Polymorphisms in Dopamine Receptor D3 Gene

DR D3 gene (DRD3) contains six exons and expresses five alternative mRNAs [49]. Among DRD3 polymorphisms, one of the most studied is located in the region coding for the receptor’s amino-terminal extracellular domain and consists in a G-to-A substitution at position 25 (rs6280) (amino acid changing Ser9Gly). This SNP has a frequency of about 40% in Caucasian population. G (Ser9) allele is referred to as allele 1, and A (Gly9) allele as allele 2. AA homozygosis confers a higher DA receptor binding affinity [50].
A clinical role was found also for rs6280 and rs1800828 SNPs [51], while neither a biological or a clinical role for the remainder SNPs in DRD3 (rs963468, rs2134655, rs9817063, rs320435, rs11721264, rs1800828, rs3773678, rs167770, rs167771, rs7633291 and rs2134655) has been identified so far.

2.4. Genetic Polymorphisms in Dopamine Receptor D4 Gene

DR D4 gene (DRD4) contains four exons. Exon 3, which encodes a cytoplasmic region of the receptor, contains a polymorphism characterized by a varying number of 48-bp repeats VNTR (variable number of tandem repeats) [52]. Nineteen different 48-bp VNTR in twenty-five different haplotypes, encoding for D4 receptors with eighteen different amino acid sequences, have been identified. Alleles with two, four and seven repetitions are the most frequently represented and may influence binding capacity to certain ligands. Besides the VNTR, a 120 bp functional duplication in the 5′ regulatory region was associated with increased transcription factor binding [53] and with reduced promoter activity for DRD4 [11,54].
Another important SNP in DRD4 is rs1800443, consisting in a T-to-G substitution (amino acid changing Val194Gly), which was associated with reduction in sensitivity to DA, absence of receptors in the high-affinity state [55] and reduced spiperone binding affinity [56]. rs1800955 consists of a T-to-C change and was associated with greater promoter activity, [57] although this result was not replicated in other studies [54,58,59]. Finally, rs747302, a SNP that consists of a C-at-G substitution at position 62 was associated with longer time to develop visual hallucinations in PD patients [18].

2.5. Genetic Polymorphisms in Dopamine Receptor D5 Gene

Genetic polymorphisms’ presence in DR D5 gene (DRD5) has been less extensively studied. Some SNPs in DRD5, including rs77434021, rs2076907, rs6283 and rs1800762, were located in both coding regions and regulatory regions, the latter including the promoter region and 5′ UTR, which may be involved in the regulation of gene expression [60]. Additionally, a G-to-T substitution (named rs6824806) has been associated with DRD5 mRNA abundance.
Finally, from a clinical point of view, only the (CA)n dinucleotide repeat, a SNP closely linked to the DRD5, has been associated with a significant risk for ADHD [61,62,63].

3. Genetic Polymorphisms in Dr and Parkinson’s Disease Risk

Genetic variation of D1-like receptor genes (rs5330, rs5331, rs13306309, rs4532) was studied in association with PD risk, but results were so far inconclusive [63]. As regards D2-like receptor genes, the role of rs1800497 (Taq1A) SNP in DRD2 on PD risk has been evaluated in several studies. McGuire et al., in a large cohort of more than 1000 patients and controls, detected significant differences among patients with different ethnic origins. In particular, non-Hispanic patients with rs1800497 homozygosis had a 1.5-fold increased risk of PD, while Afro-American patients carrying one rs1800497 allele had a 80–90% PD risk reduction. Moreover, these subjects presented a significant positive association with other DRD2 SNPs, such as rs6279 and −141 CIns/Del. Authors also hypothesized that these SNPs may be in linkage disequilibrium with other causative polymorphisms [64]. Some other studies support the positive associations between rs1800497 SNP and PD risk [65,66], however, other works do not confirm this [67,68,69,70,71,72].
Another SNP in DRD2 associated with PD risk was an intronic dinucleotide repeat; in particular, Planté-Bordeneuve showed that patients carrying a 122 bp repeat allele had an increased risk of developing PD [73]. On the contrary, other SNPs in DRD2 (such as rs1076563, rs6279, rs6278, rs273482, rs1799732, rs17294542, rs1800498, rs2234689) did not show a significant association with PD development [74].
The role of rs6280 SNP in DRD3 in PD risk is still controversial, since association with earlier PD onset was detected by one study, but not confirmed by another [75,76].
A 5′ UTR 120 bp duplication polymorphism in DRD4 was reported to influence PD risk in a cohort of Indian subjects. In particular, the 120-bp duplicated allele was protective, while the presence of only one duplicated allele predisposed to PD. When comparing patients from North and South India, the latter presented a positive association with another SNP (rs1800955) in DRD4 but, due to absence of concordant reports, no definite conclusion was stated [63]. Finally, Kronenberg analyzed a 48-bp VNTR at the third exon of the DRD4 and found an increased frequency of 4/4 genotype (that is four 48-bp VNTR on both alleles) in patients than controls, even if this difference was not statistically significant [77].

4. Genetic Polymorphisms in DR and Parkinson’s Disease Symptoms

4.1. PD Motor Symptoms

The role of DRs variation in influencing motor features of PD has been poorly studied. There is a German study on a population of 591 PD patients, exploring rs6280 SNP in DRD3, a variation previously associated with essential tremor (ET), in which no significant association between the SNP and PD tremor was detected [78,79]. As regards rigidity, there is only one report on 126 African-Caribbean patients showing a positive association with −141 C Ins/Del SNPs in DRD2, even though it was referred to antipsychotic-induced parkinsonism and not idiopathic PD [80].

4.2. Gastrointestinal Symptoms

As highlighted by the PRIAMO study, almost all PD patients suffer from gastrointestinal (GI) symptoms such as drooling of saliva, difficulty in swallowing, constipation and nausea/vomiting [81]. Some of these symptoms, like constipation, may even precede motor manifestations by several years, whereas others, like nausea and vomiting, occur early in the disease course and may represent adverse events of the pharmacological therapy [82]. GI functions are controlled by the central, autonomic and enteric nervous system, and the involvement of dopaminergic neurons has been recognized. Thus, dopaminergic neuronal loss and Lewy bodies deposition are probably involved in impaired gastric emptying that leads to GI symptoms [83,84].
Rieck et al. investigated the relationship between rs1799732 SNP in DRD2 and rs6280 SNP in DRD3 and GI symptoms induced by levodopa treatment in a group of 217 PD patients with (25.8%) or without (74.2%) GI symptoms. The authors found that both SNPs were associated with GI symptoms occurrence during levodopa therapy and that their effects were independent and probably additive [85]. Since DRD2 and DRD3 are widely distributed in the area postrema and in the gastrointestinal tract, it is conceivable that SNPs in such receptor genes influence their density and DA affinity and consequently contribute to GI symptom development [84,85].

4.3. Impulse Control Disorders

Impulse Control Disorders (ICDs) are a category of behavioral disorders characterized by a failure to resist a temptation, urge or impulse repetitively, excessively and compulsively, despite personal and relational consequences [86]. ICDs include pathological gambling, compulsive buying, binge eating, hypersexuality and other impulsive-compulsive behaviors (ICBs) such as hobbyism, punding and the Dopamine Dysregulation Syndrome (DDS) [86]. The pathophysiology of ICDs is not yet completely understood. Different studies suggest that they may be related to neurotransmitter dysfunctions, including DA, norepinephrine and serotonin. In particular, there is great interest in DA networks [86,87,88], indeed, different studies dealing with brain activity provided evidence of alterations in ventral striatum, orbito-frontal cortex and anterior cingulate cortex functioning [89]. Several risk factors for developing ICDs have been identified, including personality traits of high novelty-seeking or of impulsiveness, depression, male sex, substance abuse, younger age, younger age of PD onset, pre-PD history of ICDs, current cigarette smoking, family history of substance abuse or of ICDs, preserved executive functions, higher aggressiveness, irritability, disinhibition and eating disorders [90]. Nevertheless, the main risk factor remains DRT. ICDs affect up to 17.7% of patients with PD on DRT, whereas the prevalence of ICDs in untreated PD patients is similar to that of the general population. However, since not all patients develop this side effect, there should be individual genetic susceptibility.
For this reason, many studies have pointed out an association between genetic polymorphisms in DR and ICD.
The role of genetic polymorphisms in DRD1 in ICDs is controversial. Accordingly, while Zainal Abidin showed that SNPs rs4867798, rs4532 and rs1800497 in DRD1 were associated with an increased ICDs risk among PD patients in Malaysia [91], another two studies failed to find any association [92,93].
Moving to DRD3, Lee studied 404 Korean PD patients and 559 Korean controls and found that TT genotype of rs6280 SNP in DRD3 was independently associated with the occurrence of ICD [94]. This result is in agreement with a prospective, case-control study of Krishnamoorthy, which showed an independent association between rs6280 in DRD3 and ICDs in a cohort of 170 Indian PD patients (70 with and 100 without ICDs) and 285 healthy volunteers [95]. Finally, Castro-Martinez associated rs6280 and ICD in a sample of Spanish PD patients with an early onset of the disease (defined by an age at onset of less than 45 years old) [96]. Altogether, these data suggest that the strongest genetic risk factor for ICD development in PD patients may be rs6280. This is not surprising, as DRD3 is mainly expressed in the limbic system and it is thought to act as an auto-receptor in the ventral tegmental area, providing presynaptic negative feedback for regulating extracellular DA level. This polymorphism confers a different receptor affinity, thus impairing the reward–risk assessment in the mesolimbic system [95,96].
Finally, no correlation between SNPs in DRD2 [91,94], DRD4 and DRD5 and ICD has, so far, been found.

4.4. Visual Hallucinations

Visual hallucinations (VHs) are abnormal perceptions without a physical stimulus and they represent the most common psychotic symptoms in PD patients [97]. Moreover, VHs is associated with later onset of dementia, increased caregiver burden, nursing home placement and increased mortality [98,99]. Prevalence of VHs increases over time, reaching about 60% after 10 years [100] and 75% after 20 years from disease onset [98]. Moreover, even though VHs are predominant in PD patients, the prevalence of non-visual hallucinations (such as tactile, auditory and olfactory ones) shows a similar positive trend: in a longitudinal study, almost 70% of patients experienced multidomain (visual plus nonvisual) hallucinations by 10 years [101].
Risk factors for developing VHs are cognitive impairment, depression, motor symptoms severity, axial impairment, sleep disruption, rapid eye movement behavior sleep disorder (RBD) and visual disturbances [97,98,99]. Regarding pharmacological therapy, an increased risk of VHs was found in PD patients treated with dopaminergic agents, such as rotigotine and rasagiline. However, anti-parkinsonian drugs, although they may worsen VHs, do not represent the main causative agents for this phenomenon [97].
Nevertheless, not all PD patients on dopaminergic therapy develop VHs, thus indicating a suitable genetic predisposition. So far, only few studies have investigated the association between genetic polymorphisms in DR genes and VH in PD patients, with inconclusive results.
Goetz did not find any association between SNPs in DRD1, DRD2, DRD3 and DRD4 and VHs, but only a trend toward over-representation of the DRD3 in patients with VHs [101]. Moreover, while Wang did not detect differences in rs1800497 (DRD2), rs6280 (DRD3) and rs6283 (DRD5) frequencies in 90 Chinese PD patients with and without hallucinations [102], different results were detected in Caucasian patients. Particularly, Makoff studied the frequencies of two SNPs in DRD2 (−141 C/del, and rs1800497) and one in DRD3 (rs6280) in 155 PD patients (84 with and 71 without hallucinations). Authors found that rs1800497 SNP was associated with late-onset hallucinations [103]. Our research group demonstrated that VHs risk was more than 10 times higher in patients carrying rs686, but not rs18100497, SNP in DRD1. Moreover, we have found that patients carrying rs4532 SNP had significantly shorter time to VHs, whereas longer time to VHs was found in subjects carrying rs747302 in DRD4 [18].
The discrepancy among these studies may have different causes. First, different definitions of VHs were used. For instance, Ferrari considered only VHs requiring medical intervention [18], whereas Makoff [103] and Wang [102] considered any VHs, regardless of their severity. Second, ethnic genetic variability may be involved in the different findings between Chinese and Caucasian PD patients. Third, it is possible that other genes and neurotransmitter pathways may be involved in the pathophysiology of VHs and psychosis in PD [104].

4.5. Cognitive Decline

Cognitive decline is one of the most common nonmotor symptoms in PD, ranging from mild cognitive impairment (MCI) to dementia [105]. MCI can be present from early stages of the disease, even in 15–20% of de novo patients and there is a cumulative likelihood of developing dementia with disease progression [106]. Approximately 10% of patients present dementia within 3 years of diagnosis, and this value increases up to 46% after 10 years and 83% after 20 years [107]. Cognitive impairment in PD is characterized by executive deficits in planning, set-shifting abilities and working memory and reflects the dopaminergic loss in the dorsal striatum. This area shows connections with the dorsolateral regions of the prefrontal cortex, involved in many executive functions, whose blood flow is modulated by dopaminergic therapy [108,109]. Since not all patients develop cognitive decline, a genetic predisposition may be taken into account.
Bäckström and colleagues investigated whether rs6277 in DRD2 can affect the development of cognitive deficits in a group of 134 PD patients prospectively followed for 6–10 years. The authors found that the T/T allele in rs6277 SNP in DRD2 was associated with an increased risk of cognitive decline in PD. Specifically, this genotype significantly increased the hazard for developing cognitive decline 3.2 times in comparison with PD patients with other DRD2 genotypes, and determined poorer performances in episodic memory and attention [110].

4.6. Sleep Attacks

Sleep attacks (SA) are defined as events of overwhelming sleepiness, sometimes preceded by warning symptoms [111]. SA typically occur during different activities of daily living (i.e., eating, writing or driving a car) and last from seconds to minutes [112]. They represent a relatively common complaint among PD patients taking dopaminergic therapy, reaching a prevalence of 13%, regardless of the ethnic group, and up to 66% of the patients had daily episodes. Currently, known risk factors for SA are older age, male sex, longer disease duration, previous report of sleep disturbances, high Epworth Sleepiness Score (ESS) and dopaminergic therapy [112,113].
The role of dopaminergic transmission in sleep modulation is well recognized: dopaminergic neurons are interposed on the sleep circuit [114] and SA are more frequent in patients treated with DA agonists, particularly pramipexole (a D3 agonist), followed by ropinirole (a D2/D3 agonist) and pergolide (a D1/D2 agonist) and SA improve when dopaminergic treatments are reduced or interrupted [113]. However, association with dopaminergic treatment is still controversial, since temporal association and dose–effect relationship have not yet been fully established [115].
Since non-ergoline DA agonists have scarce affinity to the DA D1-like receptors, it has been hypothesized that alterations in dopaminergic expression mostly involve D2 receptor family. In this context, Rissling and colleagues explored the frequency of the most common SNPs in D2-like receptors genes in a cohort of 274 PD patients, stratified in two groups based on the presence or absence of sudden onset of sleep. The authors found a significant association between the rs1800497 SNP and sudden onset of sleep [116]. Of note, DRD2 plays as a presynaptic autoreceptor in the mesocorticolimbic system whose activation, therefore, might lessen the dopaminergic tone in this system, contributing to the development of daytime sleepiness. By contrast, Paus and colleagues examined 204 PD patients (102 with sleep attacks matched to 102 without sleep attacks, with the same dopaminergic treatment) and did not find any significant association between sleep attacks and 141 C del/ins, rs1800497 SNPs both in DRD2, and rs6280 SNP in DRD3. However, the authors found higher frequency of homozygosis of 48-bp VNTR with two repeats in DRD4 among patients with sleep attacks without warning signs, concluding that this allele may represent a risk factor for sleep attacks in PD patients on dopaminergic drugs [117]. These data are summarized in Table 1

5. Genetic Polymorphisms in Dr Genes and Response to Antiparkinsonian Dopaminergic Treatment

Treatment of PD is mainly based on levodopa and DA agonists, which determine restoration of basal ganglia circuitry. However, treatment may determine the development of both motor and non-motor symptoms fluctuations [131]. As shown also in previous sections of this review, several genetic polymorphisms in DR may cause a change in density and dopaminergic activity in the CSF [132]. Therefore, one may speculate that the demand for dopaminergic medication may be genetically influenced.
On this basis, Paus and colleagues tested whether rs1800497 SNP in DRD2 receptor was associated with dopaminergic treatment in a population of 503 PD patients [133]. Since no significant differences between this SNP and therapy request were detected, the authors concluded that this variation does not influence dopaminergic requirement in PD patients. Additionally, Xu and co-workers examined the impact exerted by genetic polymorphisms in DRD2 and DRD3 on daily doses of DA agonists. In particular, they investigated the association between dinucleotide short tandem repeat (Can-STR) in DRD2 and rs6280 SNPs in DRD3 and different doses of DA agonists in 168 PD patients and found that patients carrying rs6280 needed higher doses of pramipexole for effective treatment [134]. This result was confirmed by another study reporting a correlation between rs6280 and response to pramipexole [135].
Concerning the efficacy of anti-parkinsonian therapy, both rs1800497 in DRD2 and rs6280 in DRD3 have been correlated with clinical efficacy of pramipexole. Particularly, Liu and colleagues evaluated the response rates (defined as an improvement of at least 20% of the total Unified Parkinson’s Disease Rating Scale—UPDRS score) 2 months after the introduction of pramipexole in a group of 30 Chinese PD patients. The authors found that responses were significantly higher in patients homozygous for rs6280 SNP, whereas no significant association was found for rs1800497 [135].
In the ADAGIO trial, which was designed to assess the impact of rasagiline on disease progression in a large cohort (n = 692) of PD patients, rs2283265 and rs1076560 SNPs in DRD2 were associated with a significant improvement of the UPDRS score [136]. Nonetheless, a post hoc analysis of the same trial did detect a statistically significant change of UPDRS score in the delayed phase from week 36 post rasagiline therapy [137].
In a study designed to assess the impact of rs1076560 in DRD2 on motor task performance in PD patients taking L-DOPA, treatment-induced improvements were only observed in carriers of allele T, which is associated with lower D2 receptor availability, whereas patients’ homozygotes at allele G showed no performance change [138].
Genetic polymorphisms in DR genes have also been investigated with respect to therapy discontinuation. In this regard, Arbouw et al. analyzed a population of 90 PD patients treated with ropinirole or pramipexole. The authors studied 141 C Ins/Del, CA(n) STR, rs1800497 in DRD2 and rs4646996 in DRD3 and found that the absence of a 15-repetition CA(n) repeat allele in DRD2 was associated with a lower rate, while rs4646996 in DRD3 was associated with a significantly higher rate of discontinuation [139].
Finally, regarding the side effects induced by dopaminergic agents, Dos Santos found that PD patients treated with dopamine agonist and carrying rs6280 SNPs showed an increased risk of developing VHs [129].

Levodopa-Induced Dyskinesia

Levodopa administration is associated with the development of motor complications, such as motor fluctuations and Levodopa-induced dyskinesias (LIDs) [140]. By definition, motor fluctuation is a phenomenon characterized by the worsening or reappearance of motor symptoms resulting in an “off” state, usually related to low levodopa serum levels [141]. Based on clinic-pharmacological observations, different patterns of levodopa-related motor fluctuations have been described as wearing- off (which is the most common), delayed-ON, no-ON, random ON–OFF [142]. LIDs include different hyperkinetic involuntary movements (most commonly chorea and dystonia), which occur most frequently when levodopa level is high in the plasma. In particular, patients develop limb chorea and/or cranio-cervical dystonia in ON periods and painful foot dystonia during the OFF ones. Finally, diphasic dyskinesia appears immediately after and before a single dose of levodopa and comprises phasic as well as dystonic movements [143].
Levodopa dose (but not its timing of initiation) is a major risk factor for LIDs, together with younger age at PD onset, female sex and lower body weight [144,145]. Moreover, profound inter-individual heterogeneity was found in LID, thus, it is safe to assume that genetic predisposition may play a relevant role in LID appearance. However, so far, the studies focusing on genetic differences in LID lead to conflicting results.
Regarding D1-like DR, no significant association was found between genetic polymorphisms in DRD1 and DRD5 and motor complications. In particular, two case-control studies detected no correlation between SNPs in DRD1 and DRD5 and LIDs [17,119]. Nonetheless, Dos Santos suggested that rs4532 SNP in DRD1, but not rs1800497 in DRD2, may play a protector role in the occurrence of LIDs in Brazilian PD patients, by modifying DRD1 expression levels [130].
Moving to D2-like DR, Oliveri investigated whether an intronic CA(n) short tandem repeat (STR) in DRD2 was associated with peak-dose dyskinesia in a cohort of 98 Italian PD patients. The authors concluded that 13 repetitions and 14 repetitions were more expressed in non-dyskinetic patients who presented 72% risk reduction of developing peak-dose dyskinesia [119]. On the contrary, Strong found that the 14 repetitions and/or the 14/15 repetitions are a risk factor for dyskinesia development [123]. Some differences in the study design might account for these contradictory results. For example, Oliveri detected directly peak-dose dyskinesia during an acute levodopa test, while Strong relied on patient records, including all types of dyskinesias. Moreover, Oliveri compared PD patients with and without dyskinesia, whereas Strong enrolled patients with early- and late-onset dyskinesia.
Some other SNPs in DRD2 were associated with LIDs. Redenšek found that rs1799732 was associated with LIDs time occurrence after levodopa treatment [128]. Moreover, in 2001, Wang performed a case-control study, involving 40 matched pairs of Chinese PD patients, and found that rs1800497 SNP was significantly more expressed among fluctuating patients [120]. Rieck suggested that patients carrying rs2283265, rs1076560 and rs1800497 and SNPs had significantly higher LID risk [126]. Finally, Zappia showed that 13,14 CA repeats in DRD2 CAn-STR SNP was associated with decreased risk in peak-dose dyskinesia [146]. However, even for correlations between dyskinesia and SNPs in DRD2, reported results in literature are not without contradictions. For example, unlike the above reported results, in some other case-control studies, no significant association was identified between rs1800497 and rs6277 SNPs and LID or motor fluctuations [17,122,125,127,130]
Moving to DRD3, neither Wang [120] nor Kaiser [122] found any evidence of correlations between genetic polymorphisms in this gene and dyskinesia. This result was confirmed by Paus, who investigated the impact of rs6280 SNP in DRD3 on motor complications development in a cohort of 591 patients included in the gene bank of the German Competence Network on Parkinson’s disease. The authors did not detect any association between this SNP and any kind of LIDs [124]. By contrast, Lee [125] and Comi [17] described an association between rs6280 SNP in DRD3 and LIDs. Finally, Jee-Young Lee [147] found that rs6280 in DRD3 was associated with diphasic dyskinesia. Since rs6280 SNP is associated with a weaker binding affinity to DA, it has been suggested that SNPs in DRD3 may induce LID by modifying the sensitization process of the basal ganglia circuitry.
Regarding DRD4 Kaiser [122] failed to correlate any SNP in this gene with dyskinesia and/or on-off and wearing-off phenomena development and Comi et al. [17] confirmed this negative result.
Table 2 summarises the studies analysing correlations between SNPs in DA receptor and response to pharmacological treatment.

6. Role of Dr Genetic Polymorphisms in Peripheral Immunity: Possible Relevance for Pd

Besides a more “direct” action in determining clinical progression of PD and drugs response, DR SNPs may also play an indirect role by modulating the peripheral immune response, which is involved in the pathophysiology of PD [19,20,21], therefore representing a potential therapeutic target for disease modification [152].
DA is a crucial transmitter in the neuro-immune network, and dopaminergic pathways have received increasing interest in the study of adaptive immunity. DA is able to modulate activity of several immune cell subpopulations such as: T and B cells, dendritic cells, macrophages, microglia, neutrophils as well as NK cells (reviewed in [20,153]). Moreover, immune cells express all DRs, with a higher expression of D1-like receptors. Particularly, different cell sub-populations may express different receptors patterns: D1-like receptors are more represented in naïve T cells, while D2-like receptors are more expressed in memory cells [23]. Among immune cells, CD4+ T cells are specifically affected by DA, which subserve an inhibitory loop in human CD4+ CD25 high regulatory T lymphocytes. This specialized T cell subset plays a key role in the control of immune homeostasis [154] and mediates the influence exerted by dendritic cells on the differentiation of naive CD4+ T cells. In PD animal models, these regulatory cells (Treg) are able to modulate microglia differentiation and therefore influence the neurodegenerative process. Particularly, they can modulate microglia proteomics, reducing the expression of protein involved, among others, in cell metabolism and migration and protein degradation [155], and attenuate the Th17-mediated dopaminergic neuronal loss [156]. Furthermore, Treg are reduced in 6-OHDA mice model of PD and this reduction correlates with the central change in microglia profile toward a pro-inflammatory one [157].
PD patients present changes in peripheral cells expression: different studies reported a lower lymphocyte count related to a reduced number of helper CD4+ T cells and B cells [158,159]. Recently, it has been demonstrated that in a large cohort of UK subjects, a lower lymphocyte number represents an important risk factor for a subsequent diagnosis of PD [160]. Additionally, it has recently been suggested that DR SNPs could influence immune cell functions in different ways. rs4532 and rs686 in DRD1 and rs6283 in DRD5, alone and in combination, were associated with total count of lymphocytes, as well as CD3+ and CD4+ T lymphocytes, thus indicating a prevalent functional activity of D1-like receptors on these cells [23]. Moreover, SNPs in DR including rs4523 affect Treg-induced decrease of Teff cell proliferation in healthy controls [24].
PD patients display a pro-inflammatory peripheral immune phenotype, with a production of cytokines leading to Th1 differentiation [22]. Moreover, presence of circulating auto-antibodies and T cell infiltration in CNS were found in PD subjects [161]. It was suggested that infiltration and reactivation of T cells can prime microglia into a pro-inflammatory phenotype, which can in turn trigger a further detrimental response in the CNS, thus perpetuating the ongoing neurodegenerative process of PD [19]. More recently, experimental work provided evidence on the involvement of immune dysfunction in the development of both motor and non-motor symptoms of PD, including dyskinesia [162], RBD [163] and cognitive decline [164].
Altogether, these findings underline that polymorphisms in DR would likely play a role in the immune crosstalk of PD.
Although preliminary in the field of neuro-immunomodulation in PD, the above reported examples stress the importance of the possible role of genetic differences in dopaminergic modulation of immune systems in PD. In our opinion, future investigations in this field may provide a better understanding of PD pathophysiology, and eventually help the identification of new therapeutic targets for this disease [165]

7. Conclusions

All these data highlight the importance of personal genetic predisposition in the pathophysiology of PD.
Particularly, DR SNPs may be involved not only in disease development, but also in motor and non-motor complications (dyskinesia, visual hallucinations, ICD and cognitive decline), as well as in pharmacological response and side effects induced by dopaminergic agents. Furthermore, they may modulate peripheral cells expression contributing to the creation of an impaired peripheral immunity system, which is known to play a crucial role in the pathophysiology of PD [166].
DRD2 and DRD3 SNPs represent the most promising DR genetic variations in terms of biomarkers identification. Among them, DRD2 rs1800497 and DRD3 rs6280 should be tested in large cohorts of PD patients in order to better clarify their contribution in disease progression.
The evaluation of the relationship between PD progression, response to antiparkinsonian drugs and patients’ genetic profile could be useful in clinical practice since it can help in determining biomarkers for disease evolution at the time of diagnosis and personal response to pharmacological treatment. This approach will be determinant in the creation of a causative and tailored pharmacological approach which, in addition to providing benefits for patients, would reduce the management costs of therapy.

Author Contributions

C.C., M.C. and F.M. defined the topic and developed the literature search strategy together with L.M. and M.F.; L.M. and A.F. performed the literature search screening for relevant titles and abstracts, finally selecting the titles included in the review, which were cross-validated by M.F., L.M., E.C., A.V.M. wrote the first draft of the manuscript, with the exception of the paragraph dealing with pharmacogenetics of DR, which was drafted by M.F.; F.M. and A.F. drafted tables. All authors were involved in drafting the article or revising it critically for important intellectual content, and all authors approved the final version to be published. All authors agree to be accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved, and declare to have confidence in the integrity of the contributions of their co-authors. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Fondazione CARIPLO http://www.fondazionecariplo.it (accessed on 25 February 2021)-Project 2011-0504), by Fondazione UBI per Varese Onlus (grant 18/7/2017), and by the AGING PROJECT—Department of Excellence—Università del Piemonte Orientale, to the research projects brought about by the authors over the years, which allowed the development of several concepts presented and discussed in this review paper. This paper is part of the PhD program of Clinical and Experimental Medicine and Medical Humanities (University of Insubria) for LM and AF and Medical Science and Biotechnology for EC. These PhD programs and therefore this paper constitute an important part of the project Recruiting and training Physician-Scientists to empower translational research, funded by Fondazione Cariplo.

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, or in the decision to publish the results.

References

  1. Poewe, W.; Seppi, K.; Tanner, C.M.; Halliday, G.M.; Brundin, P.; Volkmann, J.; Schrag, A.-E.; Lang, A.E. Parkinson Disease. Nat. Rev. Dis. Primer 2017, 3, 17013. [Google Scholar] [CrossRef]
  2. Váradi, C. Clinical Features of Parkinson’s Disease: The Evolution of Critical Symptoms. Biology 2020, 9, 103. [Google Scholar] [CrossRef]
  3. Kumaresan, M.; Khan, S. Spectrum of non motor symptoms in Parkinson’s disease. Cureus 2021, 13, e13275. [Google Scholar] [CrossRef] [PubMed]
  4. Corrado, L.; De Marchi, F.; Tunesi, S.; Oggioni, G.D.; Carecchio, M.; Magistrelli, L.; Tesei, S.; Riboldazzi, G.; Di Fonzo, A.; Locci, C.; et al. The Length of SNCA Rep1 Microsatellite May Influence Cognitive Evolution in Parkinson’s Disease. Front. Neurol. 2018, 9, 213. [Google Scholar] [CrossRef]
  5. Beaulieu, J.-M.; Gainetdinov, R.R. The Physiology, Signaling, and Pharmacology of Dopamine receptors. Pharmacol. Rev. 2011, 63, 182–217. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Beaulieu, J.-M.; Espinoza, S.; Gainetdinov, R.R. Dopamine receptors—IUPHAR Review 13: Dopamine receptors. Br. J. Pharmacol. 2015, 172, 1–23. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Seeman, P. Chapter 1: Historical overview: Introduction to the dopamine receptors. In The Dopamine Receptors; Springer: Berlin/Heidelberg, Germany, 2009. [Google Scholar]
  8. Martel, J.C.; Gatti McArthur, S. Dopamine Receptor Subtypes, Physiology and Pharmacology: New Ligands and Concepts in Schizophrenia. Front. Pharmacol. 2020, 11, 1003. [Google Scholar] [CrossRef] [PubMed]
  9. Romanelli, R.J.; Williams, J.T.; Neve, K.A. Chapter 6: Dopamine receptor signalling: Intracellular pathways to behaviour. In The Dopamine Receptors; Springer: Berlin/Heidelberg, Germany, 2009. [Google Scholar]
  10. Tunbridge, E.M.; Narajos, M.; Harrison, C.H.; Beresford, C.; Cipriani, A.; Harrison, P.J. Which Dopamine Polymorphisms Are Functional? Systematic Review and Meta-Analysis of COMT, DAT, DBH, DDC, DRD1-5, MAOA, MAOB, TH, VMAT1, and VMAT2. Biol. Psychiatry 2019, 86, 608–620. [Google Scholar] [CrossRef] [Green Version]
  11. D’Souza, U.M.; Russ, C.; Tahir, E.; Mill, J.; McGuffin, P.; Asherson, P.J.; Craig, I.W. Functional Effects of a Tandem Duplication Polymorphism in the 5′flanking Region of the DRD4 Gene. Biol. Psychiatry 2004, 56, 691–697. [Google Scholar] [CrossRef]
  12. Howes, O.D.; McCutcheon, R.; Owen, M.J.; Murray, R.M. The Role of Genes, Stress, and Dopamine in the Development of Schizophrenia. Biol. Psychiatry 2017, 81, 9–20. [Google Scholar] [CrossRef] [Green Version]
  13. Naumovska, Z.; Nestorovska, A.K.; Filipce, A.; Sterjev, Z.; Brezovska, K.; Dimovski, A.; Suturkova, L.J. Pharmacogenetics and Antipsychotic Treatment Response. Pril. Makedon. Akad. Na Nauk. Umet. Oddelenie Za Med. Nauki 2015, 36, 53–67. [Google Scholar]
  14. Thapar, A.; O’Donovan, M.; Owen, M.J. The Genetics of Attention Deficit Hyperactivity Disorder. Hum. Mol. Genet. 2005, 14 (Suppl. 2), R275–R282. [Google Scholar] [CrossRef] [Green Version]
  15. Heiser, P.; Friedel, S.; Dempfle, A.; Konrad, K.; Smidt, J.; Grabarkiewicz, J.; Herpertz-Dahlmann, B.; Remschmidt, H.; Hebebrand, J. Molecular Genetic Aspects of Attention-Deficit/Hyperactivity Disorder. Neurosci. Biobehav. Rev. 2004, 28, 625–641. [Google Scholar] [CrossRef] [PubMed]
  16. Bhaskar, L.V.K.S.; Kumar, S.A. Polymorphisms in Genes Encoding Dopamine Signalling Pathway and Risk of Alcohol Dependence: A Systematic Review. Acta Neuropsychiatr. 2014, 26, 69–80. [Google Scholar] [CrossRef]
  17. Comi, C.; Ferrari, M.; Marino, F.; Magistrelli, L.; Cantello, R.; Riboldazzi, G.; Bianchi, M.L.E.; Bono, G.; Cosentino, M. Polymorphisms of Dopamine Receptor Genes and Risk of L-Dopa-Induced Dyskinesia in Parkinson’s Disease. Int. J. Mol. Sci. 2017, 18, 242. [Google Scholar] [CrossRef] [Green Version]
  18. Ferrari, M.; Comi, C.; Marino, F.; Magistrelli, L.; De Marchi, F.; Cantello, R.; Riboldazzi, G.; Bono, G.; Cosentino, M. Polymorphisms of Dopamine Receptor Genes and Risk of Visual Hallucinations in Parkinson’s Patients. Eur. J. Clin. Pharmacol. 2016, 72, 1335–1341. [Google Scholar] [CrossRef] [PubMed]
  19. Kustrimovic, N.; Marino, F.; Cosentino, M. Peripheral Immunity, Immunoaging and Neuroinflammation in Parkinson’s Disease. Curr. Med. Chem. 2019, 26, 3719–3753. [Google Scholar] [CrossRef]
  20. Pinoli, M.; Marino, F.; Cosentino, M. Dopaminergic Regulation of Innate Immunity: A Review. J. Neuroimmune Pharmacol. 2017, 12, 602–623. [Google Scholar] [CrossRef]
  21. Cosentino, M.; Marino, F. Adrenergic and Dopaminergic Modulation of Immunity in Multiple Sclerosis: Teaching Old Drugs New Tricks? J. Neuroimmune Pharmacol. 2013, 8, 163–179. [Google Scholar] [CrossRef]
  22. Kustrimovic, N.; Comi, C.; Magistrelli, L.; Rasini, E.; Legnaro, M.; Bombelli, R.; Aleksic, I.; Blandini, F.; Minafra, B.; Riboldazzi, G.; et al. Parkinson’s Disease Patients Have a Complex Phenotypic and Functional Th1 Bias: Cross-Sectional Studies of CD4+ Th1/Th2/T17 and Treg in Drug-Naïve and Drug-Treated Patients. J. Neuroinflammation 2018, 15, 205. [Google Scholar] [CrossRef]
  23. Cosentino, M.; Ferrari, M.; Kustrimovic, N.; Rasini, E.; Marino, F. Influence of Dopamine Receptor Gene Polymorphisms on Circulating T Lymphocytes: A Pilot Study in Healthy Subjects. Hum. Immunol. 2015, 76, 747–752. [Google Scholar] [CrossRef] [PubMed]
  24. Cosentino, M.; Kustrimovic, N.; Ferrari, M.; Rasini, E.; Marino, F. CAMP Levels in Lymphocytes and CD4+ Regulatory T-Cell Functions Are Affected by Dopamine Receptor Gene Polymorphisms. Immunology 2018, 153, 337–341. [Google Scholar] [CrossRef]
  25. Missale, C.; Nash, S.R.; Robinson, S.W.; Jaber, M.; Caron, M.G. Dopamine receptors: From Structure to Function. Physiol. Rev. 1998, 78, 189–225. [Google Scholar] [CrossRef] [Green Version]
  26. Al-Fulaij, M.A.; Ren, Y.; Beinborn, M.; Kopin, A.S. Pharmacological Analysis of Human D1 AND D2 Dopamine Receptor Missense Variants. J. Mol. Neurosci. 2008, 34, 211–223. [Google Scholar] [CrossRef] [PubMed]
  27. Grymek, K.; Łukasiewicz, S.; Faron-Góreckaa, A.; Tworzydlo, M.; Polit, A.; Dziedzicka-Wasylewska, M. Role of Silent Polymorphisms within the Dopamine D1 Receptor Associated with Schizophrenia on D1-D2 Receptor Hetero-Dimerization. Pharmacol. Rep. 2009, 61, 1024–1033. [Google Scholar] [CrossRef] [Green Version]
  28. Gingrich, J.A.; Caron, M.G. Recent Advances in the Molecular Biology of Dopamine receptors. Annu. Rev. Neurosci. 1993, 16, 299–321. [Google Scholar] [CrossRef] [PubMed]
  29. Grandy, D.K.; Litt, M.; Allen, L.; Bunzow, J.R.; Marchionni, M.; Makam, H.; Reed, L.; Magenis, R.E.; Civelli, O. The Human Dopamine D2 Receptor Gene Is Located on Chromosome 11 at Q22-Q23 and Identifies a TaqI RFLP. Am. J. Hum. Genet. 1989, 45, 778–785. [Google Scholar] [PubMed]
  30. Noble, E.P. D2 Dopamine Receptor Gene in Psychiatric and Neurologic Disorders and Its Phenotypes. Am. J. Med. Genet. Part. B Neuropsychiatr. Genet. 2003, 116B, 103–125. [Google Scholar] [CrossRef]
  31. Zhang, J.-P.; Lencz, T.; Malhotra, A.K. D2 Receptor Genetic Variation and Clinical Response to Antipsychotic Drug Treatment: A Meta-Analysis. Am. J. Psychiatry 2010, 167, 763–772. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Ritchie, T.; Noble, E.P. Association of Seven Polymorphisms of the D2 Dopamine Receptor Gene with Brain Receptor-Binding Characteristics. Neurochem. Res. 2003, 28, 73–82. [Google Scholar] [CrossRef]
  33. Pohjalainen, T.; Rinne, J.O.; Någren, K.; Lehikoinen, P.; Anttila, K.; Syvälahti, E.K.; Hietala, J. The A1 Allele of the Human D2 Dopamine Receptor Gene Predicts Low D2 Receptor Availability in Healthy Volunteers. Mol. Psychiatry 1998, 3, 256–260. [Google Scholar] [CrossRef] [PubMed]
  34. Jönsson, E.G.; Nöthen, M.M.; Grünhage, F.; Farde, L.; Nakashima, Y.; Propping, P.; Sedvall, G.C. Polymorphisms in the Dopamine D2 Receptor Gene and Their Relationships to Striatal Dopamine Receptor Density of Healthy Volunteers. Mol. Psychiatry 1999, 4, 290–296. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Thompson, J.; Thomas, N.; Singleton, A.; Piggott, M.; Lloyd, S.; Perry, E.K.; Morris, C.M.; Perry, R.H.; Ferrier, I.N.; Court, J.A. D2 Dopamine Receptor Gene (DRD2) Taq1 A Polymorphism: Reduced Dopamine D2 Receptor Binding in the Human Striatum Associated with the A1 Allele. Pharmacogenetics 1997, 7, 479–484. [Google Scholar] [CrossRef] [PubMed]
  36. Li, T.; Arranz, M.; Aitchison, K.J.; Bryant, C.; Liu, X.; Kerwin, R.W.; Murray, R.; Sham, P.; Collier, D.A. Case-Control, Haplotype Relative Risk and Transmission Disequilibrium Analysis of a Dopamine D2 Receptor Functional Promoter Polymorphism in Schizophrenia. Schizophr. Res. 1998, 32, 87–92. [Google Scholar] [CrossRef]
  37. Duan, J.; Wainwright, M.S.; Comeron, J.M.; Saitou, N.; Sanders, A.R.; Gelernter, J.; Gejman, P.V. Synonymous Mutations in the Human Dopamine Receptor D2 (DRD2) Affect MRNA Stability and Synthesis of the Receptor. Hum. Mol. Genet. 2003, 12, 205–216. [Google Scholar] [CrossRef]
  38. Hirvonen, M.M.; Lumme, V.; Hirvonen, J.; Pesonen, U.; Någren, K.; Vahlberg, T.; Scheinin, H.; Hietala, J. C957T Polymorphism of the Human Dopamine D2 Receptor Gene Predicts Extrastriatal Dopamine Receptor Availability in Vivo. Prog. Neuropsychopharmacol. Biol. Psychiatry 2009, 33, 630–636. [Google Scholar] [CrossRef]
  39. Hirvonen, M.M.; Laakso, A.; Någren, K.; Rinne, J.O.; Pohjalainen, T.; Hietala, J. C957T Polymorphism of Dopamine D2 Receptor Gene Affects Striatal DRD2 in Vivo Availability by Changing the Receptor Affinity. Synapse 2009, 63, 907–912. [Google Scholar] [CrossRef]
  40. Hirvonen, M.; Laakso, A.; Någren, K.; Rinne, J.O.; Pohjalainen, T.; Hietala, J. C957T Polymorphism of the Dopamine D2 Receptor (DRD2) Gene Affects Striatal DRD2 Availability in Vivo. Mol. Psychiatry 2004, 9, 1060–1061. [Google Scholar] [CrossRef] [Green Version]
  41. Bertolino, A.; Fazio, L.; Di Giorgio, A.; Blasi, G.; Romano, R.; Taurisano, P.; Caforio, G.; Sinibaldi, L.; Ursini, G.; Popolizio, T.; et al. Genetically Determined Interaction between the Dopamine Transporter and the D2 Receptor on Prefronto-Striatal Activity and Volume in Humans. J. Neurosci. 2009, 29, 1224–1234. [Google Scholar] [CrossRef]
  42. Moyer, R.A.; Wang, D.; Papp, A.C.; Smith, R.M.; Duque, L.; Mash, D.C.; Sadee, W. Intronic Polymorphisms Affecting Alternative Splicing of Human Dopamine D2 Receptor Are Associated with Cocaine Abuse. Neuropsychopharmacology 2011, 36, 753–762. [Google Scholar] [CrossRef]
  43. Kaalund, S.S.; Newburn, E.N.; Ye, T.; Tao, R.; Li, C.; Deep-Soboslay, A.; Herman, M.M.; Hyde, T.M.; Weinberger, D.R.; Lipska, B.K.; et al. Contrasting Changes in DRD1 and DRD2 Splice Variant Expression in Schizophrenia and Affective Disorders, and Associations with SNPs in Postmortem Brain. Mol. Psychiatry 2014, 19, 1258–1266. [Google Scholar] [CrossRef] [PubMed]
  44. Cohen, O.S.; Weickert, T.W.; Hess, J.L.; Paish, L.M.; McCoy, S.Y.; Rothmond, D.A.; Galletly, C.; Liu, D.; Weinberg, D.D.; Huang, X.-F.; et al. A Splicing-Regulatory Polymorphism in DRD2 Disrupts ZRANB2 Binding, Impairs Cognitive Functioning and Increases Risk for Schizophrenia in Six Han Chinese Samples. Mol. Psychiatry 2016, 21, 975–982. [Google Scholar] [CrossRef] [PubMed]
  45. Bertolino, A.; Taurisano, P.; Pisciotta, N.M.; Blasi, G.; Fazio, L.; Romano, R.; Gelao, B.; Lo Bianco, L.; Lozupone, M.; Di Giorgio, A.; et al. Genetically Determined Measures of Striatal D2 Signaling Predict Prefrontal Activity during Working Memory Performance. PLoS ONE 2010, 5, e9348. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Cravchik, A.; Sibley, D.R.; Gejman, P.V. Functional Analysis of the Human D2 Dopamine Receptor Missense Variants. J. Biol. Chem. 1996, 271, 26013–26017. [Google Scholar] [CrossRef] [Green Version]
  47. Zhang, Y.; Bertolino, A.; Fazio, L.; Blasi, G.; Rampino, A.; Romano, R.; Lee, M.-L.T.; Xiao, T.; Papp, A.; Wang, D.; et al. Polymorphisms in Human Dopamine D2 Receptor Gene Affect Gene Expression, Splicing, and Neuronal Activity during Working Memory. Proc. Natl. Acad. Sci. USA 2007, 104, 20552–20557. [Google Scholar] [CrossRef] [Green Version]
  48. Huang, W.; Payne, T.J.; Ma, J.Z.; Beuten, J.; Dupont, R.T.; Inohara, N.; Li, M.D. Significant Association of ANKK1 and Detection of a Functional Polymorphism with Nicotine Dependence in an African-American Sample. Neuropsychopharmacology. 2009, 34, 319–330. [Google Scholar] [CrossRef] [PubMed]
  49. Schwartz, J.C.; Levesque, D.; Martres, M.P.; Sokoloff, P. Dopamine D3 Receptor: Basic and Clinical Aspects. Clin. Neuropharmacol. 1993, 16, 295–314. [Google Scholar] [CrossRef]
  50. Lundstrom, K.; Turpin, M.P. Proposed Schizophrenia-Related Gene Polymorphism: Expression of the Ser9Gly Mutant Human Dopamine D3 Receptor with the Semliki Forest Virus System. Biochem. Biophys. Res. Commun. 1996, 225, 1068–1072. [Google Scholar] [CrossRef] [PubMed]
  51. Ishiguro, H.; Okuyama, Y.; Toru, M.; Arinami, T. Mutation and Association Analysis of the 5′ Region of the Dopamine D3 Receptor Gene in Schizophrenia Patients: Identification of the Ala38Thr Polymorphism and Suggested Association between DRD3 Haplotypes and Schizophrenia. Mol. Psychiatry 2000, 5, 433–438. [Google Scholar] [CrossRef] [Green Version]
  52. Lichter, J.B.; Barr, C.L.; Kennedy, J.L.; Van Tol, H.H.; Kidd, K.K.; Livak, K.J. A Hypervariable Segment in the Human Dopamine Receptor D4 (DRD4) Gene. Hum. Mol. Genet. 1993, 2, 767–773. [Google Scholar] [CrossRef]
  53. Ronai, Z.; Guttman, A.; Keszler, G.; Sasvari-Szekely, M. Capillary Electrophoresis Study on DNA-Protein Complex Formation in the Polymorphic 5′ Upstream Region of the Dopamine D4 Receptor (DRD4) Gene. Curr. Med. Chem. 2004, 11, 1023–1029. [Google Scholar] [CrossRef] [PubMed]
  54. Kereszturi, E.; Kiraly, O.; Csapo, Z.; Tarnok, Z.; Gadoros, J.; Sasvari-Szekely, M.; Nemoda, Z. Association between the 120-Bp Duplication of the Dopamine D4 Receptor Gene and Attention Deficit Hyperactivity Disorder: Genetic and Molecular Analyses. Am. J. Med. Genet. Part. B Neuropsychiatr. Genet. 2007, 144B, 231–236. [Google Scholar] [CrossRef]
  55. Liu, I.S.; Seeman, P.; Sanyal, S.; Ulpian, C.; Rodgers-Johnson, P.E.; Serjeant, G.R.; Van Tol, H.H. Dopamine D4 Receptor Variant in Africans, D4valine194glycine, Is Insensitive to Dopamine and Clozapine: Report of a Homozygous Individual. Am. J. Med. Genet. 1996, 61, 277–282. [Google Scholar] [CrossRef]
  56. Michealraj, K.A.; Jatana, N.; Jafurulla, M.; Narayanan, L.; Chattopadhyay, A.; Thelma, B.K. Functional Characterization of Rare Variants in Human Dopamine Receptor D4 Gene by Genotype-Phenotype Correlations. Neuroscience 2014, 262, 176–189. [Google Scholar] [CrossRef] [PubMed]
  57. Okuyama, S.; Kawashima, N.; Chaki, S.; Yoshikawa, R.; Funakoshi, T.; Ogawa, S.I.; Suzuki, Y.; Ikeda, Y.; Kumagai, T.; Nakazato, A.; et al. A Selective Dopamine D4 Receptor Antagonist, NRA0160: A Preclinical Neuropharmacological Profile. Life Sci. 1999, 65, 2109–2125. [Google Scholar] [CrossRef]
  58. Tei, S.; Mitsuhashi, H.; Ishiura, S. Data Describing the Effect of DRD4 Promoter Polymorphisms on Promoter Activity. Data Brief. 2016, 7, 1112–1117. [Google Scholar] [CrossRef] [Green Version]
  59. Simpson, J.; Vetuz, G.; Wilson, M.; Brookes, K.J.; Kent, L. The DRD4 Receptor Exon 3 VNTR and 5′ SNP Variants and MRNA Expression in Human Post-Mortem Brain Tissue. Am. J. Med. Genet. Part. B Neuropsychiatr. Genet. 2010, 153B, 1228–1233. [Google Scholar] [CrossRef]
  60. Zhao, Y.; Ding, M.; Pang, H.; Xu, X.M.; Wang, B.J. Relationship between Genetic Polymorphisms in the DRD5 Gene and Paranoid Schizophrenia in Northern Han Chinese. Genet. Mol. Res. 2014, 13, 1609–1618. [Google Scholar] [CrossRef]
  61. Daly, G.; Hawi, Z.; Fitzgerald, M.; Gill, M. Mapping Susceptibility Loci in Attention Deficit Hyperactivity Disorder: Preferential Transmission of Parental Alleles at DAT1, DBH and DRD5 to Affected Children. Mol. Psychiatry 1999, 4, 192–196. [Google Scholar] [CrossRef] [Green Version]
  62. Manor, I.; Corbex, M.; Eisenberg, J.; Gritsenkso, I.; Bachner-Melman, R.; Tyano, S.; Ebstein, R.P. Association of the Dopamine D5 Receptor with Attention Deficit Hyperactivity Disorder (ADHD) and Scores on a Continuous Performance Test (TOVA). Am. J. Med. Genet. Part. B Neuropsychiatr. Genet. 2004, 127B, 73–77. [Google Scholar] [CrossRef]
  63. Juyal, R.C.; Das, M.; Punia, S.; Behari, M.; Nainwal, G.; Singh, S.; Swaminath, P.V.; Govindappa, S.T.; Jayaram, S.; Muthane, U.B.; et al. Genetic Susceptibility to Parkinson’s Disease among South and North Indians: I. Role of Polymorphisms in Dopamine Receptor and Transporter Genes and Association of DRD4 120-Bp Duplication Marker. Neurogenetics 2006, 7, 223–229. [Google Scholar] [CrossRef]
  64. McGuire, V.; Van Den Eeden, S.K.; Tanner, C.M.; Kamel, F.; Umbach, D.M.; Marder, K.; Mayeux, R.; Ritz, B.; Ross, G.W.; Petrovitch, H.; et al. Association of DRD2 and DRD3 Polymorphisms with Parkinson’s Disease in a Multiethnic Consortium. J. Neurol. Sci. 2011, 307, 22–29. [Google Scholar] [CrossRef] [Green Version]
  65. Oliveri, R.L.; Annesi, G.; Zappia, M.; Civitelli, D.; De Marco, E.V.; Pasqua, A.A.; Annesi, F.; Spadafora, P.; Gambardella, A.; Nicoletti, G.; et al. The Dopamine D2 Receptor Gene Is a Susceptibility Locus for Parkinson’s Disease. Mov. Disord. Off. J. Mov. Disord. Soc. 2000, 15, 127–131. [Google Scholar] [CrossRef]
  66. Grevle, L.; Güzey, C.; Hadidi, H.; Brennersted, R.; Idle, J.R.; Aasly, J. Allelic Association between the DRD2 TaqI A Polymorphism and Parkinson’s Disease. Mov. Disord. Off. J. Mov. Disord. Soc. 2000, 15, 1070–1074. [Google Scholar] [CrossRef]
  67. Costa-Mallen, P.; Costa, L.G.; Smith-Weller, T.; Franklin, G.M.; Swanson, P.D.; Checkoway, H. Genetic Polymorphism of Dopamine D2 Receptors in Parkinson’s Disease and Interactions with Cigarette Smoking and MAO-B Intron 13 Polymorphism. J. Neurol. Neurosurg. Psychiatry 2000, 69, 535–537. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Kiyohara, C.; Miyake, Y.; Koyanagi, M.; Fujimoto, T.; Shirasawa, S.; Tanaka, K.; Fukushima, W.; Sasaki, S.; Tsuboi, Y.; Yamada, T.; et al. Genetic Polymorphisms Involved in Dopaminergic Neurotransmission and Risk for Parkinson’s Disease in a Japanese Population. BMC Neurol. 2011, 11, 89. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Tan, E.-K.; Tan, Y.; Chai, A.; Tan, C.; Shen, H.; Lum, S.-Y.; Fook-Cheong, S.M.C.; Teoh, M.-L.; Yih, Y.; Wong, M.-C.; et al. Dopamine D2 Receptor TaqIA and TaqIB Polymorphisms in Parkinson’s Disease. Mov. Disord. Off. J. Mov. Disord. Soc. 2003, 18, 593–595. [Google Scholar] [CrossRef] [PubMed]
  70. Pastor, P.; Muñoz, E.; Obach, V.; Martí, M.J.; Blesa, R.; Oliva, R.; Tolosa, E. Dopamine Receptor D2 Intronic Polymorphism in Patients with Parkinson’s Disease. Neurosci. Lett. 1999, 273, 151–154. [Google Scholar] [CrossRef]
  71. Dick, F.D.; De Palma, G.; Ahmadi, A.; Osborne, A.; Scott, N.W.; Prescott, G.J.; Bennett, J.; Semple, S.; Dick, S.; Mozzoni, P.; et al. Gene-Environment Interactions in Parkinsonism and Parkinson’s Disease: The Geoparkinson Study. Occup. Environ. Med. 2007, 64, 673–680. [Google Scholar] [CrossRef] [Green Version]
  72. Singh, M.; Khan, A.J.; Shah, P.P.; Shukla, R.; Khanna, V.K.; Parmar, D. Polymorphism in Environment Responsive Genes and Association with Parkinson Disease. Mol. Cell. Biochem. 2008, 312, 131–138. [Google Scholar] [CrossRef]
  73. Planté-Bordeneuve, V.; Taussig, D.; Thomas, F.; Said, G.; Wood, N.W.; Marsden, C.D.; Harding, A.E. Evaluation of Four Candidate Genes Encoding Proteins of the Dopamine Pathway in Familial and Sporadic Parkinson’s Disease: Evidence for Association of a DRD2 Allele. Neurology 1997, 48, 1589–1593. [Google Scholar] [CrossRef] [PubMed]
  74. Dai, D.; Wang, Y.; Wang, L.; Li, J.; Ma, Q.; Tao, J.; Zhou, X.; Zhou, H.; Jiang, Y.; Pan, G.; et al. Polymorphisms of DRD2 and DRD3 Genes and Parkinson’s Disease: A Meta-Analysis. Biomed. Rep. 2014, 2, 275–281. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Hassan, A.; Heckman, M.G.; Ahlskog, J.E.; Wszolek, Z.K.; Serie, D.J.; Uitti, R.J.; van Gerpen, J.A.; Okun, M.S.; Rayaprolu, S.; Ross, O.A. Association of Parkinson Disease Age of Onset with DRD2, DRD3 and GRIN2B Polymorphisms. Parkinsonism Relat. Disord. 2016, 22, 102–105. [Google Scholar] [CrossRef] [Green Version]
  76. Keeling, B.H.; Vilariño-Güell, C.; Ross, O.A.; Wszolek, Z.K.; Uitti, R.J.; Farrer, M.J. DRD3 Ser9Gly and HS1BP3 Ala265Gly Are Not Associated with Parkinson Disease. Neurosci. Lett. 2009, 461, 74–75. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Kronenberg, M.F.; Menzel, H.J.; Ebersbach, G.; Wenning, G.K.; Luginger, E.; Gollner, M.; Ransmayr, G.; Utermann, G.; Poewe, W.; Kronenberg, F. Dopamine D4 Receptor Polymorphism and Idiopathic Parkinson’s Disease. Eur. J. Hum. Genet. 1999, 7, 397–400. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Paus, S.; Gadow, F.; Kaut, O.; Knapp, M.; Klein, C.; Klockgether, T.; Wüllner, U. Tremor in Parkinson’s Disease Is Not Associated with the DRD3 Ser9Gly Polymorphism. Parkinsonism Relat. Disord. 2010, 16, 381–383. [Google Scholar] [CrossRef] [PubMed]
  79. Jeanneteau, F.; Funalot, B.; Jankovic, J.; Deng, H.; Lagarde, J.-P.; Lucotte, G.; Sokoloff, P. A Functional Variant of the Dopamine D3 Receptor Is Associated with Risk and Age-at-Onset of Essential Tremor. Proc. Natl. Acad. Sci. USA 2006, 103, 10753–10758. [Google Scholar] [CrossRef] [Green Version]
  80. Al Hadithy, A.F.; Wilffert, B.; Stewart, R.E.; Looman, N.M.; Bruggeman, R.; Brouwers, J.R.; Matroos, G.E.; van Os, J.; Hoek, H.W.; van Harten, P.N. Pharmacogenetics of Parkinsonism, Rigidity, Rest Tremor, and Bradykinesia in African-Caribbean Inpatients: Differences in Association with Dopamine and Serotonin Receptors. Am. J. Med. Genet. B Neuropsychiatr. Genet. 2008, 147B, 890–897. [Google Scholar] [CrossRef] [Green Version]
  81. Barone, P.; Antonini, A.; Colosimo, C.; Marconi, R.; Morgante, L.; Avarello, T.P.; Bottacchi, E.; Cannas, A.; Ceravolo, G.; Ceravolo, R.; et al. The PRIAMO Study: A Multicenter Assessment of Nonmotor Symptoms and Their Impact on Quality of Life in Parkinson’s Disease. Mov. Disord. Off. J. Mov. Disord. Soc. 2009, 24, 1641–1649. [Google Scholar] [CrossRef]
  82. Sánchez-Martínez, C.M.; Choreño-Parra, J.A.; Placencia-Álvarez, N.; Nuñez-Orozco, L.; Guadarrama-Ortiz, P. Frequency and Dynamics of Non-Motor Symptoms Presentation in Hispanic Patients with Parkinson Disease. Front. Neurol. 2019, 10, 1197. [Google Scholar] [CrossRef]
  83. Pfeiffer, R.F. Gastrointestinal Dysfunction in Parkinson’s Disease. Parkinsonism Relat. Disord. 2011, 17, 10–15. [Google Scholar] [CrossRef]
  84. Arinami, T.; Gao, M.; Hamaguchi, H.; Toru, M. A Functional Polymorphism in the Promoter Region of the Dopamine D2 Receptor Gene Is Associated with Schizophrenia. Hum. Mol. Genet. 1997, 6, 577–582. [Google Scholar] [CrossRef]
  85. Rieck, M.; Schumacher-Schuh, A.F.; Altmann, V.; Callegari-Jacques, S.M.; Rieder, C.R.M.; Hutz, M.H. Association between DRD2 and DRD3 Gene Polymorphisms and Gastrointestinal Symptoms Induced by Levodopa Therapy in Parkinson’s Disease. Pharmacogenomics J. 2018, 18, 196–200. [Google Scholar] [CrossRef] [PubMed]
  86. Hellstrand, M.; Danielsen, E.A.; Steen, V.M.; Ekman, A.; Eriksson, E.; Nilsson, C.L. The Ser9gly SNP in the Dopamine D3 Receptor Causes a Shift from CAMP Related to PGE2 Related Signal Transduction Mechanisms in Transfected CHO Cells. J. Med. Genet. 2004, 41, 867–871. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Marques, A.; Durif, F.; Fernagut, P.-O. Impulse Control Disorders in Parkinson’s Disease. J. Neural Transm. 2018, 125, 1299–1312. [Google Scholar] [CrossRef] [PubMed]
  88. Cormier, F.; Muellner, J.; Corvol, J.-C. Genetics of Impulse Control Disorders in Parkinson’s Disease. J. Neural Transm. 2013, 120, 665–671. [Google Scholar] [CrossRef] [PubMed]
  89. Vargas, A.P.; Cardoso, F.E.C. Impulse Control and Related Disorders in Parkinson’s Disease. Arq. Neuropsiquiatr. 2018, 76, 399–410. [Google Scholar] [CrossRef]
  90. Santangelo, G.; Raimo, S.; Cropano, M.; Vitale, C.; Barone, P.; Trojano, L. Neural Bases of Impulse Control Disorders in Parkinson’s Disease: A Systematic Review and an ALE Meta-Analysis. Neurosci. Biobehav. Rev. 2019, 107, 672–685. [Google Scholar] [CrossRef]
  91. Latella, D.; Maggio, M.G.; Maresca, G.; Saporoso, A.F.; Le Cause, M.; Manuli, A.; Milardi, D.; Bramanti, P.; De Luca, R.; Calabrò, R.S. Impulse Control Disorders in Parkinson’s Disease: A Systematic Review on Risk Factors and Pathophysiology. J. Neurol. Sci. 2019, 398, 101–106. [Google Scholar] [CrossRef]
  92. Zainal Abidin, S.; Tan, E.L.; Chan, S.-C.; Jaafar, A.; Lee, A.X.; Abd Hamid, M.H.N.; Abdul Murad, N.A.; Pakarul Razy, N.F.; Azmin, S.; Ahmad Annuar, A.; et al. DRD and GRIN2B Polymorphisms and Their Association with the Development of Impulse Control Behaviour among Malaysian Parkinson’s Disease Patients. BMC Neurol. 2015, 15, 59. [Google Scholar] [CrossRef] [Green Version]
  93. Vallelunga, A.; Flaibani, R.; Formento-Dojot, P.; Biundo, R.; Facchini, S.; Antonini, A. Role of Genetic Polymorphisms of the Dopaminergic System in Parkinson’s Disease Patients with Impulse Control Disorders. Parkinsonism Relat. Disord. 2012, 18, 397–399. [Google Scholar] [CrossRef]
  94. Kraemmer, J.; Smith, K.; Weintraub, D.; Guillemot, V.; Nalls, M.A.; Cormier-Dequaire, F.; Moszer, I.; Brice, A.; Singleton, A.B.; Corvol, J.-C. Clinical-Genetic Model Predicts Incident Impulse Control Disorders in Parkinson’s Disease. J. Neurol. Neurosurg. Psychiatry 2016, 87, 1106–1111. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Lee, J.-Y.; Lee, E.K.; Park, S.S.; Lim, J.-Y.; Kim, H.J.; Kim, J.S.; Jeon, B.S. Association of DRD3 and GRIN2B with Impulse Control and Related Behaviors in Parkinson’s Disease. Mov. Disord. 2009, 24, 1803–1810. [Google Scholar] [CrossRef] [PubMed]
  96. Krishnamoorthy, S.; Rajan, R.; Banerjee, M.; Kumar, H.; Sarma, G.; Krishnan, S.; Sarma, S.; Kishore, A. Dopamine D3 Receptor Ser9Gly Variant Is Associated with Impulse Control Disorders in Parkinson’s Disease Patients. Parkinsonism Relat. Disord. 2016, 30, 13–17. [Google Scholar] [CrossRef] [PubMed]
  97. Castro-Martínez, X.H.; García-Ruiz, P.J.; Martínez-García, C.; Martínez-Castrillo, J.C.; Vela, L.; Mata, M.; Martínez-Torres, I.; Feliz-Feliz, C.; Palau, F.; Hoenicka, J. Behavioral Addictions in Early-Onset Parkinson Disease Are Associated with DRD3 Variants. Parkinsonism Relat. Disord. 2018, 49, 100–103. [Google Scholar] [CrossRef]
  98. Kurita, A.; Koshikawa, H.; Akiba, T.; Seki, K.; Ishikawa, H.; Suzuki, M. Visual Hallucinations and Impaired Conscious Visual Perception in Parkinson Disease. J. Geriatr. Psychiatry Neurol. 2020, 33, 377–385. [Google Scholar] [CrossRef] [PubMed]
  99. Diederich, N.J.; Fénelon, G.; Stebbins, G.; Goetz, C.G. Hallucinations in Parkinson Disease. Nat. Rev. Neurol. 2009, 5, 331–342. [Google Scholar] [CrossRef]
  100. Ravina, B.; Marder, K.; Fernandez, H.H.; Friedman, J.H.; McDonald, W.; Murphy, D.; Aarsland, D.; Babcock, D.; Cummings, J.; Endicott, J.; et al. Diagnostic Criteria for Psychosis in Parkinson’s Disease: Report of an NINDS, NIMH Work Group. Mov. Disord. Off. J. Mov. Disord. Soc. 2007, 22, 1061–1068. [Google Scholar] [CrossRef]
  101. Frei, K.; Truong, D.D. Hallucinations and the Spectrum of Psychosis in Parkinson’s Disease. J. Neurol. Sci. 2017, 374, 56–62. [Google Scholar] [CrossRef]
  102. Goetz, C.G.; Stebbins, G.T.; Ouyang, B. Visual plus Nonvisual Hallucinations in Parkinson’s Disease: Development and Evolution over 10 Years. Mov. Disord. Off. J. Mov. Disord. Soc. 2011, 26, 2196–2200. [Google Scholar] [CrossRef]
  103. Wang, J.; Zhao, C.; Chen, B.; Liu, Z.-L. Polymorphisms of Dopamine Receptor and Transporter Genes and Hallucinations in Parkinson’s Disease. Neurosci. Lett. 2004, 355, 193–196. [Google Scholar] [CrossRef]
  104. Makoff, A.J.; Graham, J.M.; Arranz, M.J.; Forsyth, J.; Li, T.; Aitchison, K.J.; Shaikh, S.; Grünewald, R.A. Association Study of Dopamine Receptor Gene Polymorphisms with Drug-Induced Hallucinations in Patients with Idiopathic Parkinson’s Disease. Pharmacogenetics 2000, 10, 43–48. [Google Scholar] [CrossRef] [PubMed]
  105. Lenka, A.; Arumugham, S.S.; Christopher, R.; Pal, P.K. Genetic Substrates of Psychosis in Patients with Parkinson’s Disease: A Critical Review. J. Neurol. Sci. 2016, 364, 33–41. [Google Scholar] [CrossRef] [PubMed]
  106. O’Callaghan, C.; Lewis, S.J.G. Cognition in Parkinson’s Disease. Int. Rev. Neurobiol. 2017, 133, 557–583. [Google Scholar]
  107. Aarsland, D.; Kurz, M.W. The Epidemiology of Dementia Associated with Parkinson Disease. J. Neurol. Sci. 2010, 289, 18–22. [Google Scholar] [CrossRef] [PubMed]
  108. Williams-Gray, C.H.; Mason, S.L.; Evans, J.R.; Foltynie, T.; Brayne, C.; Robbins, T.W.; Barker, R.A. The CamPaIGN Study of Parkinson’s Disease: 10-Year Outlook in an Incident Population-Based Cohort. J. Neurol. Neurosurg. Psychiatry 2013, 84, 1258–1264. [Google Scholar] [CrossRef] [Green Version]
  109. O’Callaghan, C.; Bertoux, M.; Hornberger, M. Beyond and below the Cortex: The Contribution of Striatal Dysfunction to Cognition and Behaviour in Neurodegeneration. J. Neurol. Neurosurg. Psychiatry 2014, 85, 371–378. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  110. Cools, R.; Stefanova, E.; Barker, R.A.; Robbins, T.W.; Owen, A.M. Dopaminergic Modulation of High-Level Cognition in Parkinson’s Disease: The Role of the Prefrontal Cortex Revealed by PET. Brain J. Neurol. 2002, 125, 584–594. [Google Scholar] [CrossRef] [Green Version]
  111. Bäckström, D.; Eriksson Domellöf, M.; Granåsen, G.; Linder, J.; Mayans, S.; Elgh, E.; Zetterberg, H.; Blennow, K.; Forsgren, L. Polymorphisms in Dopamine-Associated Genes and Cognitive Decline in Parkinson’s Disease. Acta Neurol. Scand. 2018, 137, 91–98. [Google Scholar] [CrossRef]
  112. Homann, C.N.; Wenzel, K.; Suppan, K.; Ivanic, G.; Kriechbaum, N.; Crevenna, R.; Ott, E. Sleep Attacks in Patients Taking Dopamine Agonists: Review. BMJ 2002, 324, 1483–1487. [Google Scholar] [CrossRef] [Green Version]
  113. Paus, S.; Brecht, H.M.; Köster, J.; Seeger, G.; Klockgether, T.; Wüllner, U. Sleep Attacks, Daytime Sleepiness, and Dopamine Agonists in Parkinson’s Disease. Mov. Disord. Off. J. Mov. Disord. Soc. 2003, 18, 659–667. [Google Scholar] [CrossRef]
  114. Yeung, E.Y.H.; Cavanna, A.E. Sleep Attacks in Patients with Parkinson’s Disease on Dopaminergic Medications: A Systematic Review. Mov. Disord. Clin. Pract. 2014, 1, 307–316. [Google Scholar] [CrossRef] [Green Version]
  115. Monti, J.M.; Monti, D. The Involvement of Dopamine in the Modulation of Sleep and Waking. Sleep Med. Rev. 2007, 11, 113–133. [Google Scholar] [CrossRef]
  116. Rissling, I.; Geller, F.; Bandmann, O.; Stiasny-Kolster, K.; Körner, Y.; Meindorfner, C.; Krüger, H.-P.; Oertel, W.H.; Möller, J.C. Dopamine Receptor Gene Polymorphisms in Parkinson’s Disease Patients Reporting “Sleep Attacks”. Mov. Disord. Off. J. Mov. Disord. Soc. 2004, 19, 1279–1284. [Google Scholar] [CrossRef] [PubMed]
  117. Paus, S.; Seeger, G.; Brecht, H.M.; Köster, J.; El-Faddagh, M.; Nöthen, M.M.; Klockgether, T.; Wüllner, U. Association Study of Dopamine D2, D3, D4 Receptor and Serotonin Transporter Gene Polymorphisms with Sleep Attacks in Parkinson’s Disease. Mov. Disord. Off. J. Mov. Disord. Soc. 2004, 19, 705–707. [Google Scholar] [CrossRef] [PubMed]
  118. Goetz, C.G.; Burke, P.F.; Leurgans, S.; Berry-Kravis, E.; Blasucci, L.M.; Raman, R.; Zhou, L. Genetic Variation Analysis in Parkinson Disease Patients with and without Hallucinations: Case-Control Study. Arch. Neurol. 2001, 58, 209–213. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Oliveri, R.L.; Annesi, G.; Zappia, M.; Civitelli, D.; Montesanti, R.; Branca, D.; Nicoletti, G.; Spadafora, P.; Pasqua, A.A.; Cittadella, R.; et al. Dopamine D2 Receptor Gene Polymorphism and the Risk of Levodopa-Induced Dyskinesias in PD. Neurology 1999, 53, 1425–1430. [Google Scholar] [CrossRef] [PubMed]
  120. Wang, J.; Liu, Z.L.; Chen, B. Dopamine D5 Receptor Gene Polymorphism and the Risk of Levodopa-Induced Motor Fluctuations in Patients with Parkinson’s Disease. Neurosci. Lett. 2001, 308, 21–24. [Google Scholar] [CrossRef]
  121. Wang, J.; Liu, Z.L.; Chen, B. Association Study of Dopamine D2, D3 Receptor Gene Polymorphisms with Motor Fluctuations in PD. Neurology 2001, 56, 1757–1759. [Google Scholar] [CrossRef]
  122. Kaiser, R.; Hofer, A.; Grapengiesser, A.; Gasser, T.; Kupsch, A.; Roots, I.; Brockmöller, J. L -Dopa-Induced Adverse Effects in PD and Dopamine Transporter Gene Polymorphism. Neurology 2003, 60, 1750–1755. [Google Scholar] [CrossRef]
  123. Strong, J.A.; Dalvi, A.; Revilla, F.J.; Sahay, A.; Samaha, F.J.; Welge, J.A.; Gong, J.; Gartner, M.; Yue, X.; Yu, L. Genotype and Smoking History Affect Risk of Levodopa-Induced Dyskinesias in Parkinson’s Disease. Mov. Disord. Off. J. Mov. Disord. Soc. 2006, 21, 654–659. [Google Scholar] [CrossRef]
  124. Paus, S.; Gadow, F.; Knapp, M.; Klein, C.; Klockgether, T.; Wüllner, U. Motor Complications in Patients Form the German Competence Network on Parkinson’s Disease and the DRD3 Ser9Gly Polymorphism. Mov. Disord. Off. J. Mov. Disord. Soc. 2009, 24, 1080–1084. [Google Scholar] [CrossRef]
  125. Lee, J.-Y.; Cho, J.; Lee, E.-K.; Park, S.-S.; Jeon, B.S. Differential Genetic Susceptibility in Diphasic and Peak-Dose Dyskinesias in Parkinson’s Disease. Mov. Disord. Off. J. Mov. Disord. Soc. 2011, 26, 73–79. [Google Scholar] [CrossRef] [PubMed]
  126. Rieck, M.; Schumacher-Schuh, A.F.; Altmann, V.; Francisconi, C.L.; Fagundes, P.T.; Monte, T.L.; Callegari-Jacques, S.M.; Rieder, C.R.; Hutz, M.H. DRD2 Haplotype Is Associated with Dyskinesia Induced by Levodopa Therapy in Parkinson’s Disease Patients. Pharmacogenomics 2012, 13, 1701–1710. [Google Scholar] [CrossRef] [PubMed]
  127. Kaplan, N.; Vituri, A.; Korczyn, A.D.; Cohen, O.S.; Inzelberg, R.; Yahalom, G.; Kozlova, E.; Milgrom, R.; Laitman, Y.; Friedman, E.; et al. Sequence Variants in SLC6A3, DRD2, and BDNF Genes and Time to Levodopa-Induced Dyskinesias in Parkinson’s Disease. J. Mol. Neurosci. MN 2014, 53, 183–188. [Google Scholar] [CrossRef] [PubMed]
  128. Redenšek, S.; Jenko Bizjan, B.; Trošt, M.; Dolžan, V. Clinical-Pharmacogenetic Predictive Models for Time to Occurrence of Levodopa Related Motor Complications in Parkinson’s Disease. Front. Genet. 2019, 10, 461. [Google Scholar] [CrossRef]
  129. Dos Santos, E.U.D.; Duarte, E.B.C.; Miranda, L.M.R.; Asano, A.G.C.; Asano, N.M.J.; Maia, M. de M.D.; de Souza, P.R.E. Influence of DRD1 and DRD3 Polymorphisms in the Occurrence of Motor Effects in Patients with Sporadic Parkinson’s Disease. Neuromolecular Med. 2019, 21, 295–302. [Google Scholar] [CrossRef] [PubMed]
  130. Dos Santos, E.U.D.; Sampaio, T.F.; Tenório Dos Santos, A.D.; Bezerra Leite, F.C.; da Silva, R.C.; Crovella, S.; Asano, A.G.C.; Asano, N.M.J.; de Souza, P.R.E. The Influence of SLC6A3 and DRD2 Polymorphisms on Levodopa-Therapy in Patients with Sporadic Parkinson’s Disease. J. Pharm. Pharmacol. 2019, 71, 206–212. [Google Scholar] [CrossRef]
  131. Chaudhuri, K.R.; Rizos, A.; Sethi, K.D. Motor and Nonmotor Complications in Parkinson’s Disease: An Argument for Continuous Drug Delivery? J. Neural Transm. Vienna Austria 1996 2013, 120, 1305–1320. [Google Scholar] [CrossRef] [Green Version]
  132. Noble, E.P. The D2 Dopamine Receptor Gene: A Review of Association Studies in Alcoholism and Phenotypes. Alcohol Fayettev. N. 1998, 16, 33–45. [Google Scholar] [CrossRef]
  133. Paus, S.; Grünewald, A.; Klein, C.; Knapp, M.; Zimprich, A.; Janetzky, B.; Möller, J.C.; Klockgether, T.; Wüllner, U. The DRD2 TaqIA Polymorphism and Demand of Dopaminergic Medication in Parkinson’s Disease. Mov. Disord. Off. J. Mov. Disord. Soc. 2008, 23, 599–602. [Google Scholar] [CrossRef]
  134. Xu, S.; Liu, J.; Yang, X.; Qian, Y.; Xiao, Q. Association of the DRD2 CAn-STR and DRD3 Ser9Gly Polymorphisms with Parkinson’s Disease and Response to Dopamine Agonists. J. Neurol. Sci. 2017, 372, 433–438. [Google Scholar] [CrossRef] [PubMed]
  135. Liu, Y.-Z.; Tang, B.-S.; Yan, X.-X.; Liu, J.; Ouyang, D.-S.; Nie, L.-N.; Fan, L.; Li, Z.; Ji, W.; Hu, D.-L.; et al. Association of the DRD2 and DRD3 Polymorphisms with Response to Pramipexole in Parkinson’s Disease Patients. Eur. J. Clin. Pharmacol. 2009, 65, 679–683. [Google Scholar] [CrossRef] [PubMed]
  136. Masellis, M.; Collinson, S.; Freeman, N.; Tampakeras, M.; Levy, J.; Tchelet, A.; Eyal, E.; Berkovich, E.; Eliaz, R.E.; Abler, V.; et al. Dopamine D2 Receptor Gene Variants and Response to Rasagiline in Early Parkinson’s Disease: A Pharmacogenetic Study. Brain J. Neurol. 2016, 139, 2050–2062. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Bhattacharjee, S.; Hughes, E.; Ng, K.L. Rasagiline Sensitive Dopamine D2 Receptor Gene Variants: A Step Forward Toward More Personalized Antiparkinsonian Therapy. Mov. Disord. Clin. Pract. 2017, 4, 181–182. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Kwak, Y.; Bohnen, N.I.; Müller, M.L.T.M.; Dayalu, P.; Burke, D.T.; Seidler, R.D. Task-Dependent Interactions between Dopamine D2 Receptor Polymorphisms and L-DOPA in Patients with Parkinson’s Disease. Behav. Brain Res. 2013, 245, 128–136. [Google Scholar] [CrossRef]
  139. Arbouw, M.E.L.; Movig, K.L.L.; Egberts, T.C.G.; Poels, P.J.E.; van Vugt, J.P.P.; Wessels, J.A.M.; van der Straaten, R.J.H.M.; Neef, C.; Guchelaar, H.-J. Clinical and Pharmacogenetic Determinants for the Discontinuation of Non-Ergoline Dopamine Agonists in Parkinson’s Disease. Eur. J. Clin. Pharmacol. 2009, 65, 1245–1251. [Google Scholar] [CrossRef] [Green Version]
  140. Chaudhuri, K.R.; Jenner, P.; Antonini, A. Should There Be Less Emphasis on Levodopa-induced Dyskinesia in Parkinson’s Disease? Mov. Disord. 2019, 34, 816–819. [Google Scholar] [CrossRef] [PubMed]
  141. Marsden, C.D.; Parkes, J.D. “On-off” Effects in Patients with Parkinson’s Disease on Chronic Levodopa Therapy. Lancet 1976, 1, 292–296. [Google Scholar] [CrossRef]
  142. Chou, K.L.; Stacy, M.; Simuni, T.; Miyasaki, J.; Oertel, W.H.; Sethi, K.; Fernandez, H.H.; Stocchi, F. The Spectrum of “off” in Parkinson’s Disease: What Have We Learned over 40 Years? Parkinsonism Relat. Disord. 2018, 51, 9–16. [Google Scholar] [CrossRef]
  143. Bastide, M.F.; Meissner, W.G.; Picconi, B.; Fasano, S.; Fernagut, P.-O.; Feyder, M.; Francardo, V.; Alcacer, C.; Ding, Y.; Brambilla, R.; et al. Pathophysiology of L-Dopa-Induced Motor and Non-Motor Complications in Parkinson’s Disease. Prog. Neurobiol. 2015, 132, 96–168. [Google Scholar] [CrossRef] [PubMed]
  144. Cilia, R.; Akpalu, A.; Sarfo, F.S.; Cham, M.; Amboni, M.; Cereda, E.; Fabbri, M.; Adjei, P.; Akassi, J.; Bonetti, A.; et al. The Modern Pre-Levodopa Era of Parkinson’s Disease: Insights into Motor Complications from Sub-Saharan Africa. Brain J. Neurol. 2014, 137, 2731–2742. [Google Scholar] [CrossRef] [Green Version]
  145. Picillo, M.; Palladino, R.; Barone, P.; Erro, R.; Colosimo, C.; Marconi, R.; Morgante, L.; Antonini, A. PRIAMO Study Group the PRIAMO Study: Urinary Dysfunction as a Marker of Disease Progression in Early Parkinson’s Disease. Eur. J. Neurol. 2017, 24, 788–795. [Google Scholar] [CrossRef]
  146. Zappia, M.; Annesi, G.; Nicoletti, G.; Arabia, G.; Annesi, F.; Messina, D.; Pugliese, P.; Spadafora, P.; Tarantino, P.; Carrideo, S.; et al. Sex Differences in Clinical and Genetic Determinants of Levodopa Peak-Dose Dyskinesias in Parkinson Disease: An Exploratory Study. Arch. Neurol. 2005, 62, 601–605. [Google Scholar] [CrossRef] [PubMed]
  147. Kakinuma, S.; Beppu, M.; Sawai, S.; Nakayama, A.; Hirano, S.; Yamanaka, Y.; Yamamoto, T.; Masafumi, C.; Aisihaer, X.; Aersilan, A.; et al. Monoamine Oxidase B Rs1799836 G Allele Polymorphism Is a Risk Factor for Early Development of Levodopa-Induced Dyskinesia in Parkinson’s Disease. eNeurologicalSci 2020, 19, 100239. [Google Scholar] [CrossRef]
  148. Greenbaum, L.; Goldwurm, S.; Zozulinsky, P.; Lifschytz, T.; Cohen, O.S.; Yahalom, G.; Cilia, R.; Tesei, S.; Asselta, R.; Inzelberg, R.; et al. Do Tardive Dyskinesia and L-Dopa Induced Dyskinesia Share Common Genetic Risk Factors? An Exploratory Study. J. Mol. Neurosci. 2013, 51, 380–388. [Google Scholar] [CrossRef] [PubMed]
  149. Kvernmo, T.; Härtter, S.; Burger, E. A Review of the Receptor-Binding and Pharmacokinetic Properties of Dopamine Agonists. Clin. Ther. 2006, 28, 1065–1078. [Google Scholar] [CrossRef] [PubMed]
  150. Scheller, D.; Ullmer, C.; Berkels, R.; Gwarek, M.; Lübbert, H. The in Vitro Receptor Profile of Rotigotine: A New Agent for the Treatment of Parkinson’s Disease. Naunyn. Schmiedebergs Arch. Pharmacol. 2009, 379, 73–86. [Google Scholar] [CrossRef] [PubMed]
  151. Damasceno Dos Santos, E.U.; Duarte, E.B.C.; Miranda, L.M.R.; Asano, A.G.C.; Asano, N.M.J.; Maia, M.D.M.D.; de Souza, P.R.E. Pharmacogenetic Profile and the Occurrence of Visual Hallucinations in Patients with Sporadic Parkinson’s Disease. J. Clin. Pharmacol. 2019, 59, 1006–1013. [Google Scholar] [CrossRef]
  152. Gendelman, H.E.; Zhang, Y.; Santamaria, P.; Olson, K.E.; Schutt, C.R.; Bhatti, D.; Shetty, B.L.D.; Lu, Y.; Estes, K.A.; Standaert, D.G.; et al. Evaluation of the Safety and Immunomodulatory Effects of Sargramostim in a Randomized, Double-Blind Phase 1 Clinical Parkinson’s Disease Trial. NPJ Park. Dis. 2017, 3, 10. [Google Scholar] [CrossRef]
  153. Cappellano, G.; Carecchio, M.; Fleetwood, T.; Magistrelli, L.; Cantello, R.; Dianzani, U.; Comi, C. Immunity and inflammation in neurodegenerative diseases. Am. J. Neurodegener Dis. 2013, 2, 89–107. [Google Scholar]
  154. Cosentino, M.; Fietta, A.M.; Ferrari, M.; Rasini, E.; Bombelli, R.; Carcano, E.; Saporiti, F.; Meloni, F.; Marino, F.; Lecchini, S. Human CD4+CD25+ Regulatory T Cells Selectively Express Tyrosine Hydroxylase and Contain Endogenous Catecholamines Subserving an Autocrine/Paracrine Inhibitory Functional Loop. Blood 2007, 109, 632–642. [Google Scholar] [CrossRef] [Green Version]
  155. Reynolds, A.D.; Stone, D.K.; Mosley, R.L.; Gendelman, H.E. Proteomic studies of nitrated alpha-synuclein microglia regulation by CD4+CD25+ T cells. J. Proteome Res. 2009, 8, 3497–3511. [Google Scholar] [CrossRef] [Green Version]
  156. Reynolds, A.D.; Stone, D.K.; Hutter, J.A.; Benner, E.J.; Mosley, R.L.; Gendelman, H.E. Regulatory T cells attenuate Th17 cell-mediated nigrostriatal dopaminergic neurodegeneration in a model of Parkinson’s disease. J. Immunol. 2010, 184, 2261–2271. [Google Scholar] [CrossRef] [Green Version]
  157. Ambrosi, G.; Kustrimovic, N.; Siani, F.; Rasini, E.; Cerri, S.; Ghezzi, C.; Dicorato, G.; Caputo, S.; Marino, F.; Cosentino, M.; et al. Complex Changes in the Innate and Adaptive Immunity Accompany Progressive Degeneration of the Nigrostriatal Pathway Induced by Intrastriatal Injection of 6-Hydroxydopamine in the Rat. Neurotox. Res. 2017, 32, 71–81. [Google Scholar] [CrossRef]
  158. Baba, Y.; Kuroiwa, A.; Uitti, R.J.; Wszolek, Z.K.; Yamada, T. Alterations of T-lymphocyte populations in Parkinson disease. Parkinsonism Relat. Disord. 2005, 11, 493–498. [Google Scholar] [CrossRef]
  159. Stevens, C.H.; Rowe, D.; Morel-Kopp, M.C.; Orr, C.; Russell, T.; Ranola, M.; Ward, C.; Halliday, G.M. Reduced T helper and B lymphocytes in Parkinson’s disease. J. Neuroimmunol. 2012, 252, 95–99. [Google Scholar] [CrossRef] [Green Version]
  160. Sarkar, S.; Dammer, E.B.; Malovic, E.; Olsen, A.L.; Raza, S.A.; Gao, T.; Xiao, H.; Oliver, D.L.; Duong, D.; Joers, V.; et al. Molecular Signatures of Neuroinflammation Induced by ASynuclein Aggregates in Microglial Cells. Front. Immunol. 2020, 11, 33. [Google Scholar] [CrossRef]
  161. Contaldi, E.; Magistrelli, L.; Milner, A.V.; Cosentino, M.; Marino, F.; Comi, C. Expression of transcription factors in CD4+ T cells as potential biomarkers of motor complications in Parkinson’s disease. J. Parkinsons Dis. 2020, 26, 1–8. [Google Scholar] [CrossRef]
  162. De Francesco, E.; Terzaghi, M.; Storelli, E.; Magistrelli, L.; Comi, C.; Legnaro, M.; Mauri, M.; Marino, F.; Versino, M.; Cosentino, M. CD4+ T-Cell Transcription Factors in Idiopathic REM Sleep Behavior Disorder and Parkinson’s Disease. Mov. Disord. Off. J. Mov. Disord. Soc. 2021, 36, 225–229. [Google Scholar] [CrossRef]
  163. Magistrelli, L.; Storelli, E.; Rasini, E.; Contaldi, E.; Comi, C.; Cosentino, M.; Marino, F. Relationship between Circulating CD4+ T Lymphocytes and Cognitive Impairment in Patients with Parkinson’s Disease. Brain. Behav. Immun. 2020, 89, 668–674. [Google Scholar] [CrossRef] [PubMed]
  164. Magistrelli, L.; Comi, C. Beta2-Adrenoceptor Agonists in Parkinson’s Disease and Other Synucleinopathies. J. Neuroimmune Pharmacol. 2020, 15, 74–81. [Google Scholar] [CrossRef] [PubMed]
  165. Prado, C.; Contreras, F.; González, H.; Díaz, P.; Elgueta, D.; Barrientos, M.; Herrada, A.A.; Lladser, Á.; Bernales, S.; Pacheco, R. Stimulation of Dopamine Receptor D5 Expressed on Dendritic Cells Potentiates Th17-Mediated Immunity. J. Immunol. 2012, 188, 3062–3070. [Google Scholar] [CrossRef]
  166. Franz, D.; Contreras, F.; González, H.; Prado, C.; Elgueta, D.; Figueroa, C.; Pacheco, R. Dopamine receptors D3 and D5 Regulate CD4(+)T-Cell Activation and Differentiation by Modulating ERK Activation and CAMP Production. J. Neuroimmunol. 2015, 284, 18–29. [Google Scholar] [CrossRef]
Table 1. Studies concerning effects of single nucleotide polymorphisms (SNPs) in dopaminergic receptors (DR) in Parkinson’s disease (PD) patients.
Table 1. Studies concerning effects of single nucleotide polymorphisms (SNPs) in dopaminergic receptors (DR) in Parkinson’s disease (PD) patients.
Study PopulationConsidered SNPsMain ResultsReference
Parkinson Disease (PD) Risk
100 PD patients and 100 healthy controlsDRD2: dinucleotide (GT) repeat Individuals who were homozygous for allele 3 were more frequent in the sporadic PD than in controls.[73]
122 PD patients and 127 healthy controlsDRD4: VNTR 7/48-base pair repeatNo association with PD risk.[77]
154 PD patients and 125 healthy controlsDRD2: TG/CA repeat (intron 2)No association with PD risk.[70]
152 PD patients and 231 healthy controlsDRD2: rs1800497 (2137 C > T)
rs1079597 (54716 G > A)
No association with PD risk.[67]
135 PD patients and 202 healthy controlsDRD2: rs1800497 (2137 C > T)
rs1079597 (54716 G > A)
rs1801028 (−141 C ins/del)
rs1799732 (932 C > G)
rs1800497 (CC genotype) and rs1079597 (GG genotype) were more frequent in PD patients than in healthy controls.[65]
72 PD patients and 81 healthy controlsDRD2: rs1800497 (2137 C > T)rs1800497 (CC genotype) was more frequent in PD patients than in healthy controls.[66]
204 PD patients and 216 healthy controlsDRD2: rs1800497 (2137 C > T)
rs1079597 (54716 G > A)
No association with PD risk.[69]
487 PD patients and 474 healthy controlsDRD1: rs4532 (−48 A > G)
rs5330 (150 G > T)
rs5331 (595 T > G)
rs13306309 (685 G > A)
DRD2: rs1799732 (−141 C ins/del)
rs1800497 (2137 C > T)
rs1800498 (59414 C > T)
rs2234689 (72519 C > G)
DRD3: rs6280 (25 A > G)
rs324026 (32213 G > A)
rs1503670 (23948 C > G)
rs905568 (g.773 G > A)
DRD4: VNTR 120 bp duplication
−521 C > T
VNTR 7/48-base pair repeat
DRD4 12-bp duplication seems to be associated with PD risk.[63]
767 PD patients and 1989 healthy controlsDRD2: rs1800497 (2137 C > T)
rs1079597 (54716 G > A)
No association with PD risk.[71]
70 PD patients and 100 healthy controlsDRD2: rs1800497 (2137 C > T)
rs1079597 (54716 G > A)
No association with PD risk.[72]
448 PD patients and 428 healthy controlsDRD3: rs6280 (25 A > G)No association with PD risk.[76]
1325 PD patients and 1735 healthy controlsDRD2: rs1800497 (2137 C > T)
rs1076563 (55093 T > G)
rs6279 (376 C > G)
rs6278 (725 G > T)
rs273482 (18232490 C > T)
DRD3: rs6280 (25 A > G)
rs2134655 (65054 G > A)
DRD2 rs1800497 (TT genotype) was associated with increased PD risk in non-Hispanic whites.
DRD3 rs6280 (GG genotype) was associated with a decreased PD risk in Hispanics.
[64]
293 PD patients and 369 healthy controlsDRD2: rs1800497 (2137 C > T)
DRD4: rs1800955 (4480 T > G)
No association with PD risk.[68]
4279 PD patients and 5661 healthy controlsDRD2: rs1800497 (2137 C > T)
rs1079597 (54716 G > A)
rs6279 (376 C > G)
rs6278 (725 G > T)
rs273482 (18232490 C > T)
rs1799732 (−141 C ins/del)
rs1076563 (55093 T > G)
DRD3: rs6280 (25 A > G)
rs2134655 (65054 G > A)
DRD3 rs2134655 and DRD2 rs1800497 were associated with PD risk.[74]
664 PD patients and 718 healthy controlsDRD2: rs1800497 (2137 C > T)
DRD3: rs6280 (25 A > G)
DRD3 rs6280 (genotype CC) was associated with earlier age at onset.[75]
PD Motor Symptoms
126 PD patients (31 with tremor)DRD2 rs1799732 (−141 ins/del)
DRD2 rs6280
DRD2 −141 ins/del was associated with rigidity.[80]
591 PD patients (62 with tremor)DRD3 rs6280 (25 A > G)No association with tremor in PD.[78]
Gastrointestinal Symptoms
217 PD patients (56 with GS)DRD2: rs1799732 (−141 C ins/del)
DRD3: rs6280 (25 G > A)
rs1799732 (−141 C ins/ins genotype) and DRD3 rs6280 (GG genotype) was associated with levodopa-induced gastrointestinal symptoms.[85]
Impulse Control Disorders (ICD)
404 PD patients (58 with ICD) and 559 healthy controlsDRD2: rs1800497 (2137 C > T)
DRD3: rs6280 (25 G > A)
DRD3 rs6280 (AA genotype) was associated with ICD.[95]
89 PD patients (41 with ICDs)DRD2: rs1800497 (2137 C > T)No association with ICD.[93]
91 PD patients (52 with ICD)DRD1: rs4532 (−48 A > G)
rs4867798 (863 A > C)
rs265981 (−684 T > A)
DRD2: rs1800497 (2137 C > T)
DRD3: rs6280 (25 G > A)
rs3732783 (51 A > G)
DRD1 rs4532 (AG genotype), rs4867798 (CC genotype) and DRD2 rs1800497 (TT genotype) were associated with ICD increased risk.[92]
276 PD patients (52 with ICD)DRD2: rs1800497 (2137 C > T)
DRD3: rs6280 (25 G > A)
No association with ICD.[94]
170 PD patients (70 with ICD) and 285 healthy controls DRD3: rs6280 (25 G > A)DRD3 rs6280 (CT genotype) is associated with ICD.[96]
126 non early-onset PD (NEOPD) and 73 EOPD (age at onset < 45).DRD3: rs6280 (25 G > A)rs6280 was associated with ICD in PD patients with an early onset of the disease.[97]
Visual Hallucinations (VHs)
155 PD patients (84 with VHs)DRD2 rs1800497 2137 C > T
rs1799732 (141 C ins/del)
DRD3: rs6280 (25 G > A)
rs1800497 (C allele) was associated with late-onset hallucination.[104]
88 PD patients (44 with chronic hallucinations)DRD1 B1/B2
DRD2 rs1801028 (932 C > G)
DRD3 1/2
DRD4 7/48-base pair repeat (exon 3)
No association with VHs.[118]
90 PD patients (45 with VHs)DRD2: rs1800497 (2137 C > T)
DRD3: rs6280 (25 G > A)
DRD5: rs6283 (978 T > C)
No association with VHs.[103]
82 PD patients (42 with VHs)DRD1: rs4532 (−48 A > G)
rs686 (62 G > A)
DRD2: rs1800497 2137 C > T
rs6277 (957 C > T)
DRD3: rs6280 (25 G > A)
rs1800828 (−712 G > C)
DRD4: rs747302 (−616 C > G)
VNTR (7/48-base pair repeat)
DRD5: rs6283 (978 T > C)
DRD1 rs686 (AA genotype) was associated with increased VHs risk.
DRD1 rs4532 (GG genotype) and rs686 (TT genotype) displayed shorter time to VHs, whereas a longer time to VHs was found in subjects carrying DRD4 rs747302 (CG genotypes).
[18]
Cognitive Decline
134 patients (84 with dementia)DRD2 rs6277 (957 C > T)T/T genotype was associated with increased risk of developing dementia.[111]
Sleep Attacks
274 PD patients, 137 with sudden onset of sleep (SOS)DRD2: rs1800497 2137 C > T
DRD3: rs6280 (25 G > A)
DRD4: VNTR (7/48-base pair repeat)
DRD2 rs1800497 (allele C) was associated with SOS.[116]
204 PD patients (102 with sleep attacks)DRD2: rs1799732 (141 C ins/del)
DRD2: rs1800497 (2137 C > T)
DRD3: rs6280 (25 G > A)
DRD4: VNTR (7/48-base pair repeat)
DRD4*2 (short) allele was associated with sleep attacks without warning signs. [117]
Motor Complications
136 PD patients and 224 healthy controlsDRD1 D1.1 in the 5′ untranslated region (UTR), D1.7 in the 39 UTR
DRD2 short tandem repeat polymorphism (STRP)
STRP 13- and 14-repetition genotype polymorphism were significantly higher in non-dyskinetic PD patients compared with dyskinetic patients.[119]
140 PD patients and 140 healthy controlsDRD2: rs1800497 (2137 C > T)
DRD3: Bal1, Msp1
DRD2 rs1800497 SNPs may be associated with increased risk for motor fluctuations in PD.[120]
120 PD patients and 110 healthy controlsDRD5 978 T > C No association with risk of developing motor fluctuation.[121]
183 PD patientsDRD2: rs1800497 (2137 C > T) rs1800496 (928 C > T)
rs1801028 67518 C > G
DRD3: rs6280 (25 A > G)
DRD4: VNTR (7/48-base pair repeat)
No association with L-Dopa induced ADR.[122]
92 PD patientsDRD2 short tandem repeat polymorphism (STRP)STRP 14- and/or 14/15-repetition genotype was a risk factor for dyskinesia.[123]
591 PD patientsDRD3: rs6280 (25 A > G)No association with motor complications.[124]
503 PD patients and 559 healthy controlsDRD2: rs1800497 (2137 C > T)
DRD3: rs6280 (25 A > G)
DRD3 rs6280 (AA genotype) was associated with diphasic dyskinesia.[125]
199 PD patientsDRD2: rs1800497 (2137 C > T)
rs1799732 (−141 C ins/del)
rs2283265 (67314 G > T)
rs6277 (957 C > T)
rs1076560 (63314 G > T)
rs2734849 (1472 A > G)
DRD2 rs2283265, rs1076560, rs100497 are associated with dyskinesia. [126]
352 PD patientsDRD2: rs4245147 (32995 G > A)
rs6275 (939 T > C)
rs6276 (52 G > A)
rs6277 (957 C > T)
rs4630328 (16793 C > T)
rs17529477 (33935 C > T)
rs1079597 (54716 G > A)
rs4938017 (47073 G > A)
rs4245148 (30583 A > C)
rs1079594 (68190 T > A)
rs1800497 (2137 C > T)
No association with LID[127]
100 PD patients (50 with dyskinesia)DRD1: rs4532 (−48 A > G)
rs686 (62 G > A)
DRD2: rs1800497 2137 C > T
rs6277 (957 C > T)
DRD3: rs6280 (25 G > A)
rs1800828 (−712 G > C)
DRD4: rs747302 (−616 C > G)
VNTR (7/48-base pair repeat)
DRD5: rs6283 (978 T > C)
DRD3 rs2680 was associated with earlier LID development. [17]
220 PD patientsDRD2 rs1799732DRD2 rs1799732 was associated with LIDs. [128]
228 PD patientsDRD1 rs4532 (−48 A > G)
DRD3 rs6280 (25 A > G)
DRD1 rs4532 (G/G genotype) may play a role in occurrence of LID.[129]
195 PD patientsDRD2 rs1800497 (2137 C > T)No association with the development of motor fluctuations/dyskinesia.[130]
Table 2. Studies concerning effects of SNPs in DA receptor on response to antiparkinsonian drug therapy.
Table 2. Studies concerning effects of SNPs in DA receptor on response to antiparkinsonian drug therapy.
Approved IndicationsReceptor Affinity
(ki, nm)
Ddd (g)Adm.rSnpsMain ResultsRefs.
Levodopa
PD [b]Dopamine precursor with no intrinsic receptor affinity, which acts indirectly by increasing dopamine levels [b]3.5 [a]O [a]DRD1 rs4532
DRD3 rs6280
DRD1 was associated with occurrence of LID.[129]
DRD1 rs4532
DRD2 rs1799732 rs1800497 rs1079597
DRD3 rs6280
No associations between tested SNPs and LID [147]
DRD2 rs1799732 rs1801028 DRD3 rs6280DRD2 rs1799732 and DRD3 rs6280 were associated to occurrence of motor fluctuation after levodopa treatment. DRD2 rs1799732 was associated with time occurrence of LID [128]
DRD2 rs1800497No associations with motor fluctuation or LID[130]
DRD1 rs4532 rs686
DRD2 rs1800497 2137 rs6277
DRD3 rs6280 rs1800828
DRD4 rs747302
DRD5 rs6283
DRD3 rs2680 was associated with earlier LID[17]
DRD2 rs1799732 DRD3 rs6280DRD2 rs1799732 and DRD3 rs6280 were associated with an increased prevalence of gastrointestinal symptoms associated with levodopa treatment[85]
DRD2: rs4245147 rs6275 rs6276, rs6277 rs4630328 rs17529477 rs1079597 rs4938017 rs4245148 rs1079594 rs1800497No association with LID[127]
DRD3 rs6280
DRD2 rs1800497
DRD3 rs6280 and DRD2 rs1800497 were not associated with LID in Israel and Italian sample of PD[148]
DRD2 rs 1076560DRD2 rs1076560 was not associated with motor sequence learning task and change in manual motor abilities after levodopa treatment[138]
DRD2 rs2283265 rs1076560 rs6277, rs1800497, rs2734849DRD2 rs2283265, rs1076560, rs100497 were associated with LID[126]
DRD2 rs1800497
DRD3 rs6280
DRD3 rs6280 (AA genotype) was associated with diphasic, but not pick-dose LID[125]
DRD3 rs6280DRD3 rs6280 did not increase susceptibility to develop levodopa-induced motor complications [133]
DRD2 short tandem repeat polymorphism (STRP)STRP 14 and/or 14/15 repetition genotype was a risk factor for LID[123]
DRD2 CAn-STR13,14 CA repeats was associated with decreased risk to developing peak-dose LID [146]
DRD2 rs1800497 rs1800496
rs1801028
DRD3 rs6280
DRD4 VNTR (7/48-base pair repeat)
No association with ADR L-Dopa induced[122]
DRD5 rs6283No association with risk of developing motor fluctuation during levodopa treatment[121]
DRD2 STR polymorphism 13 and 14 alleles of the DRD2 STR SNP was correlated with risk reduction of developing peak-dose LID[119]
Pergolide
PD [b]D2 (0.2); D3 (0.5); D4 (1.3); D5 (164); D1 (172)
Others: 5HT1A (1.9); 5HT2A (8.3); 5HT1D (13.2); 5HT2B (7.1); 5HT1B (281.8); 5HT2C (295.1) [149]
0.003 [a]O [a]DRD2 rs1799732, rs1800497 and DRD3 rs6280D2DR rs1800497 was associated with late-onset VHs, but not with early dopamine-induced VHs [104]
Ropinirole
PD and RLS [b]D3 (2.9); D2 (3.7); D4 (7.8), D5 (41,211); D1 (36,600) Others: 5HT1A (288); 5HT1 B (>10,000); 5HT1 D (1380); 5HT2A (>10,000); 5HT2B (3802); 5HT2C (>10,000) [149]0.006 [a]O [a]DRD2 rs1799732, (CA)n STR, rs1800497, DRD3 MscIThe absence of a 15× DRD2 CA repeat allele was related to a decreased discontinuation of ropinirole treatment[139]
Pramipexole
PD [b]D3 (0.5); D2 (3.9); D4 (5.1), D5 (>10,000); D1 (>50,000)
Others: 5HT1A (692); 5HT1 B (8318); 5HT1 D (1660); 5HT2A (>10,000); 5HT2B (>10,000); 5HT2C (>10,000) [149]
0.0025 [a]O [a]DRD3 rs6280DRD3 rs6280 showed a significant pattern of interaction upon behavioural addiction and higher doses for effective treatment[97,122,123]
DRD3 rs6280DRD3 rs6280 (G/G genotype) require higher pramipexole doses for effective treatment[134]
DRD3 rs6280DRD3 rs6280 (A/A genotype) was associate with response rate to pramipexole[135]
Rotigotine
PD and RLS [b]D3 (0.71); D4 (3.9–15); D5 (5.4); D2 (13.5); D1 (83) Others: AR- α1A (176); AR- α1B (273); AR- α2A (338); AR- α2B (27); AR- α2C (135); 5HT1A (30); 5HT7 (86); H1 (330) [150]0.006 [a]TD [a]DRD1 rs4532
DRD3 rs6280
DRD3 rs6280 resulted to increased prevalence for VHs[151]
Abbreviations. DDD, Defined Daily Dose; ADM.R, administration route; NA: Not Available; O, oral administration, P, parenteral, S-Q, subcutaneous, SL, sublingual, IV, intravenous, TD, transdermal; LID, L-dopa Induced Dyskinesia; VHs, Visual Hallucinations; PD, Parkinson Diseases; ADR, Adverse Drug Reactions; COMT, Catechol-O-methyltransferase; MAO, monoamine oxidase; NMDA, N-methyl-D-aspartate receptor; RLS, restless legs syndrome. [a] https://www.whocc.no (accessed on 25 February 2021); [b] https://www.guidetopharmacology.org/ (accessed on 25 February 2021).
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Magistrelli, L.; Ferrari, M.; Furgiuele, A.; Milner, A.V.; Contaldi, E.; Comi, C.; Cosentino, M.; Marino, F. Polymorphisms of Dopamine Receptor Genes and Parkinson’s Disease: Clinical Relevance and Future Perspectives. Int. J. Mol. Sci. 2021, 22, 3781. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22073781

AMA Style

Magistrelli L, Ferrari M, Furgiuele A, Milner AV, Contaldi E, Comi C, Cosentino M, Marino F. Polymorphisms of Dopamine Receptor Genes and Parkinson’s Disease: Clinical Relevance and Future Perspectives. International Journal of Molecular Sciences. 2021; 22(7):3781. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22073781

Chicago/Turabian Style

Magistrelli, Luca, Marco Ferrari, Alessia Furgiuele, Anna Vera Milner, Elena Contaldi, Cristoforo Comi, Marco Cosentino, and Franca Marino. 2021. "Polymorphisms of Dopamine Receptor Genes and Parkinson’s Disease: Clinical Relevance and Future Perspectives" International Journal of Molecular Sciences 22, no. 7: 3781. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22073781

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

Article Metrics

Back to TopTop