IMR Press / FBL / Volume 27 / Issue 9 / DOI: 10.31083/j.fbl2709265
Open Access Original Research
Single Nucleotide Polymorphisms of Indoleamine 2,3-Dioxygenase 1 Influenced the Age Onset of Parkinson's Disease
Show Less
1 ELKH-SZTE Neuroscience Research Group, Danube Neuroscience Research Laboratory, Eötvös Loránd Research Network, University of Szeged (ELKH-SZTE), Danube Neuroscience Research Laboratory, H-6725 Szeged, Hungary
2 Institute of Biophysics, Biological Research Centre, Eötvös Loránd Research Network (ELKH), H-6726 Szeged, Hungary
3 Department of Neurology and Cerebrovascular Diseases, Pándy Kálmán County Hospital, H-5700 Gyula, Hungary
4 Department of Medical Microbiology and Immunobiology, Albert Szent-Györgyi Medical School, University of Szeged, H-6725 Szeged, Hungary
5 Department of Medical Physics and Informatics, Albert Szent-Györgyi Medical School, University of Szeged, H-6720 Szeged, Hungary
6 Department of Neurology, Albert Szent-Györgyi Medical School, University of Szeged, H-6725 Szeged, Hungary
*Correspondence: vecsei.laszlo@med.u-szeged.hu (László Vécsei)
These authors contributed equally.
Academic Editor: Graham Pawelec
Front. Biosci. (Landmark Ed) 2022, 27(9), 265; https://doi.org/10.31083/j.fbl2709265
Submitted: 11 August 2022 | Revised: 6 September 2022 | Accepted: 13 September 2022 | Published: 27 September 2022
Copyright: © 2022 The Author(s). Published by IMR Press.
This is an open access article under the CC BY 4.0 license.
Abstract

Background: Earlier studies reported alterations of the kynurenine (KYN) pathway of tryptophan (TRP) metabolism in Parkinson’s disease (PD). The first rate-limiting enzymes indoleamine 2,3-dioxygenase (IDO) and tryptophan dioxygenase were observed upregulated, resulting elevated KYN/TRP ratios in the serum and cerebrospinal fluid samples of patients with PD. More and more single nucleotide polymorphisms (SNPs) have been identified in a population of PD. However, little is known about the impact of genetic variations of the IDO on the pathogenesis of PD. Methods: SNP analysis of IDO1 was performed by allelic discrimination assay with fluorescently labelled TaqMan probes and a subgroup analysis was conducted according to the age of PD onset. The frame shifts variant rs34155785, intronic variant rs7820268, and promotor region variant rs9657182 SNPs of 105 PD patients without comorbidity were analyzed and compared to 129 healthy controls. Results: No significant correlation was found in three SNPs between PD patients and healthy controls. However, the subgroup analysis revealed that A alleles of rs7820268 SNP or rs9657182 SNP carriers contribute to later onset of PD than non-carriers. Conclusions: The study suggested that SNPs of IDO1 influenced the age onset of PD and genotyping of SNPs in certain alleles potentially serves as a risk biomarker of PD.

Keywords
kynurenine
tryptophan
indoleamine 2
3-dioxygenase
IDO
Parkinson's diseases
single nucleotide polymorphisms
biomarker
age
onset
1. Introduction

Parkinson’s disease (PD) is the second most prevalent chronic progressive neurodegenerative disease characterized by motor symptoms such as tremor, rigidity, and hypokinesia. The prevalence of PD is approximately 0.2% on average in the general population, but it is increasing with age up to 1.9% [1]. The disease affects primarily the elderly, imposing a serious burden on the aging societies. The non-motor symptoms may appear in early stages of the disease and even before the appearance of classical motor symptoms [2]. The comorbidity is common in PD patients [3, 4, 5, 6, 7, 8]. Relatively significant findings are autonomic dysfunction including arrhythmia, blood pressure irregularity, asymmetric sweating, and incontinence and psychobehavioral manifestations including dementia, depression, anxiety, paranoia, and psychosis [9, 10]. The occurrence of dysfunctions in autonomic modulation in PD patients mostly impacts on psychobehavioral responses, and usually overlaps with behavioral disorders [11, 12, 13, 14, 15]. The histopathological and clinical hallmarks of PD include the degeneration of dopaminergic neurons in the substantia nigra pars compacta, the presence of Lewy bodies, and the positive response to dopamine (DA) replacement therapy [16, 17].

The etiology of PD remains poorly understood. Genetic disposition, neurodevelopmental insuts, and environmental factors are considered to play an important role in the pathogenesis of PD [18]. Pathognomic findings are abnormal protein aggregation, elevated oxidative stress, mitochondrial dysfunction, increased glutamate excitotoxicity, alteration of immune response, disturbance of the kynurenine pathway (KP) of tryptophan (TRP) metabolism, and among others [19, 20, 21, 22, 23, 24, 25, 26, 27]. Altered levels and ratios of kynurenine (KYN) metabolites have been observed in neurologic and psychiatric diseases; furthermore, KYN metabolites have attracted a growing attention not only as potential biomarkers, but also as cross-species and environmenental indicators [10, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36]. Earlier studies revealed that the activities of indoleamine 2,3-dioxygenase (IDO) 1/TRP 2,3-dioxygenase (TDO) were upregulated in PD patients compared to controls, which was indicated by elevation of L-KYN/TRP ratios in the serum and in cerebrospinal fluid (CSF) samples of the patients [37]. The cascade of the KP produces several neuroactive metabolites such as 3-hydroxykynurenine (3-HK) quinolinic acid (QUIN), 3-hydroxyanthranillic acid (3-HAA), and kynurenic acid (KYNA) [38].

3-HK and 3-HAA generate reactive oxygen species (ROS) [16]. An elevation of 3-HK levels was related to excitotoxic injury and is observed in patients with neurodegenerative diseases [10]. The neurotoxic effects of 3-HK and 3-HAA involve the generation of superoxide anion and hydrogen peroxide, which contribute to the oxidative processes implicated in the pathophysiology of meningitis [39]. QUIN is a free-radical metabolite. Interferon (IFN)-γ activates IDO, formamidase, and kynurenine 3-monooxygenase (KMO) activities in human microglial cells and macrophages, increasing QUIN synthesis [40].

KYNA is a broad-spectrum, competitive antagonist of all three ionotropic excitatory glutamate receptors including α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor, kainate receptor, and N-methyl-D-aspartate (NMDA) receptor [41]. However, KYNA exhibited dual actions at AMPA receptors dependent on its concentration: inhibitory at micromolar concentrations, while excitatory at nanomolar concentrations. The micromolar concentrations are inhibitory, while the nanomolar concentrations are facilitatory by allosteric modulation of the AMPA receptor [42, 43]. Furthermore, KYNA facilitates memory enhancement in low doses [44]. The actions of KYNA at the α-7 nicotinic acetylcholine receptor remain controversial [45]. KYNA has antioxidant properties that scavenges ROS to suppress overshooting inflammation in damaging tissues. Reduced concentrations of KYNA may contribute to tissue damage and inflammatory cell proliferation neurodegenerative diseases [10, 16, 46]. In addition, KYNA was reported to contribute to weight reduction without affecting osteogenesis [46].

The indoleamine 2,3-dioxygenase IDO1 is one of the first rate-limiting enzymes that converts L-TRP to N-formyl KYN in TRP metabolism, which play a crucial role in governing concentrations of downward bioactive KYN metabolites [16] (Fig. 1). Activation of IDO1 and the KYN system promotes immunosuppressive effects by inhibition of Natural Killer cells, inhibition of T cell functions, and activation of the regulatory T cells [47]. The relationship between PD and the KP have been investigated in in vivo and in vitro studies; however, only limited data are available on the significance of genetic alterations of KP enzymes in PD patients [48, 49].

Fig. 1.

The kynurenine pathway (KP). The KP is the degradation route of tryptophan (TRP) metabolism producing an end-product nicotinamide adenine dinucleotide (NAD). The indoleamine 2,3-dioxygenase (IDO) 1, IDO2, and the tryptophan 2,3-dioxygenase (TDO) (1) are the first rate-liming enzymes that convert the L-tryptophan (TRP) to N-formyl-L-kynurenine. N-formyl-L-kynurenine is converted by formamidase (2) to L-kynurenine (L-KYN). L-KYN is metabolized into various bioactive compounds: kynurenic acid, picolinic acid, 3-hydroxy-L-kynurenine (3-HK), and quinolinic acid. The following are main enzymes of the KP: 1: tryptophan dioxygenase (TDO) and indoleamine 2,3-dioxygenase (IDO), 2: formamidase, 3: kynurenine aminotransferases (KATs), 4: kynurenine 3-monooxygenase (KMO), 5: kynureninase, 6: kynureninase, 7: non-specific hydroxylation, 8: 2-amino-3-carboxymuconate-semialdehyde decarboxylase (ACMSD), 9: 3-hydroxyanthranilic acid oxygenase, 10: quinolinic acid phosphoribosyltransferase.

In this study we performed single nucleotide polymorphisms (SNPs) analysis in three loci of IDO1 in PD patients and healthy controls. The frameshift mutation rs34155785 SNP causes a drastic change in the gene product. Both intronic variant rs7820268 SNP, and promotor region variant rs9657182 SNP affect the immune system. The T allelic variants of the rs7820268 showed impaired CD8+ regulatory T cell function, while the C allele of rs9657182 SNP was found more susceptible to IFN-alpha treatment, which induced depressive symptoms [50] (Fig. 2). Secondly, a subgroup analysis was conducted according to the age of PD onset in search of the genetic link between the IDO1 variants and PD.

Fig. 2.

Schematic of the indoleamine 2,3-dioxygenase (IDO) 1 gene. The IDO1 gene consist of 10 exons and 9 introns localized in 8p11.21 chromosome region. It has 9 transcript variants. The encoded protein is 403 amino acid long. The rs9657182 SNP is located 5’ upstream from the promoter region of the IDO1 gene. The rs7820268 SNP is localized in the fourth intron and the last rs34155785 SNP, which is a frame shift mutation, is located in the sixth exon.

2. Materials and Methods
2.1 Ethics

The blood samples of 105 PD patients and 129 healthy controls were examined for the study. All patients gave their informed consent in accordance with the Declaration of Helsinki, and the study was approved by the Medical Research Council Scientific and Research Ethics Committee (47066-3/2013/EKU (556/2013)). The samples were collected at two sites (at the Department of Neurology, Faculty of Medicine, University of Szeged, Hungary, and at the Department of Neurology and Cerebrovascular Diseases, Pándy Kálmán County Hospital, Gyula, Hungary). The patient and the control groups were age- and gender-matched (gender ratio p = 0.908 by chi-square test; mean age difference p = 0.310 by Student’s t-test). For data analysis, the patient’s group was divided into two subgroups based on the appearance of the first symptoms: the early-onset (EOPD; disease onset 60 years) and the late-onset (LOPD; disease onset >60 years) group. The onset of the disease could not be established for two patients. The sociodemographic data of the two groups are presented in Table 1.

Table 1.The sociodemographic data of PD patients and controls.
Group Male Female Age Age at onset (Mean ± SD)
Mean ± SD Median Min Max
PD patients (n = 105) 48 57 66.42 ± 9.24 68 34 84 58.81 ± 10.970
Controls (n = 129) 58 71 65.26 ± 8.1 63 53 87 -
Min, minimum age in the group; Max, maximum age in the group.
2.2 DNA Isolation and Polymerase Chain Reaction

Peripheral whole blood samples (stored at –80 °C) were subjected to genomic DNA isolation by the desalting method developed by Miller et al. [51]. The purified genomic DNA were stored at –20 °C at the biobank of the Department of Neurology, Faculty of Medicine, University of Szeged (biobank license: Regional Human Biomedical Research Ethics Committee: 135/2008).

The IDO1 genotypes were determined by allelic discrimination study with TaqMan probes. Three SNP of the IDO1 gene were investigated. The rs34155785 SNP is a frame shift mutation in the human genome (results a Phe (F) >Leu (L) exchange), which localize in the sixth exon of the IDO1 gene. The rs7820268 SNP is intronic SNP (G/A change) of the IDO1 gene is localized in the fourth intron. The rs9657182 SNP (G/A change) is localized 5’ upstream from the beginning of the IDO1 promoter region.

For the rs34155785 SNP, the following primers were used for the amplification of the DNA forward primer: 5’- CTA AAC TTC TTG CCT TCC TTA TC-3’; reverse primer: 5’- AGA CGT ACT TTG ATT GCA GA-3’. The following probes were applied for allelic discrimination: wild type allele: 5’-Fam- GAC GTT TTG TTC TCA TTT CGT G-BHQ-1-3’; and C allele: 5’-Hex- GAC GTT TTG TTG CTC ATT TCG TG-BHQ-1-3’.

The rs7820268 SNP, the following primers were applied for the amplification of the G/A at chromosome 8: forward primer: 5’- TAA ATG TAA TGC CTA CTG AAG AA-3’; reverse primer: 5’- CCT TAT GAA AGC AGC CAT G-3’. The following probes were designed for allelic discrimination: G allele: 5’-Fam- GTA GCA TTC AAT CAA ATA GCA ACA AC-1-3’; and A allele: 5’-Hex- GTA GCA TTC AAT TAA ATA GCA ACA AC-1-3’.

The rs9657182 SNP, the following primers were used: forward primer: 5’- ATT GTT GTA GGT CAT AAA AGG AG-3’; reverse primer: 5’- TGA AGA CAC AAC ACT TAA GGA-3’. The following probes were applied for the separation of the alleles: G allele: 5’-Fam- CCA TCT TTA ACC ACG GCC A-BHQ-1-3’; A allele: 5’-Hex- CCA TCT TTA ACC ATG GCC A-1-3’.

The parameters for PCR amplifications were as follows: 95 °C for 3 min, followed by 44 cycles of 95 °C for 10 s, and then 59 °C for 50 s (rs34155785 SNP) or 57 °C for 50 s (rs7820268 SNP) or 55 °C for 50 s (rs9657182 SNP). The genotyping specific master mix from the PCR Biosystem (2x PCRBio Genotyping mix Lo-ROX) was used. The PCR experiments were performed with a Bio-Rad CFX96 C1000 real-time thermal cycler machine, and the data analysis was carried out with Bio-Rad CFX Manager version 1.6 (Applied Biosystems, University Park, IL, USA).

2.3 Statistical Methods

For the analysis of genotype frequencies and allele distributions SPSS software version 26.0 (IBM Corp., Chicago, IL, USA) and GraphPad Prism 6.01 (GraphPad Software, San Diego, CA, USA) was used. We applied the chi-square test for comparing the distributions of the examined genotypes and alleles, and the t-test for comparing the averages in the two groups. Odds ratio (OR) and 95% confidence interval (CI) was calculated for the SNP variants in the groups of early and late onset. A p value less than 0.05 was considered statistically significant.

3. Results

We determined the frequencies of three polymorphisms of IDO1 gene in PD patients and matching healthy controls. The observed genotype frequencies of the PD and the control groups were in accordance with the Hardy-Weinberg equilibrium.

The genotype distribution of rs34155785 was 105 (100%) homozygous wild-types in the PD patient group, and 129 (100%) in the control group, we did not detect the insertion allele in either group. The allele frequencies were 210 (100%) wild-type alleles in the patient and 258 (100%) in the control group. This SNP variant is probably not associated with PD and do not affect the age at disease onset either, as no allelic differences could be observed between the two groups.

The genotype distribution of rs7820268 in the patient cohort was 56 GG, 38 GA and 11 AA, and in the control group was 59 GG, 63 GA and 7 AA. The allele frequencies were very similar, with 71.4% G allele and 28.6% A allele in the PD group and 70.2% G and 29.8% A in the controls (Table 2). This SNP variant is not significantly associated with the PD (genotype: p = 0.093, G allele: p = 0.149, A allele: p = 0.248), but it affects the age at disease onset (genotype: p = 0.042). The frequency of A allele was significantly higher in LOPD group than in EOPD group (p = 0.024, OR = 2.471 95% CI (1.117–5.467)) (Table 3).

Table 2.Genotype and allele distribution of the rs7820268 and rs9657182 SNPs.
rs7820268
Genotype PD patients Controls χ2 p value
GG 56 (53.3%) 59 (45.7%) 4.744 (df = 2) 0.098
AG 38 (36.2%) 63 (48.8%)
AA 11 (10.5%) 7 (5.4%)
Allele
G 150 (71.4%) 181 (70.2%) 0.091 (df = 1) 0.763
A 60 (28.6%) 77 (29.8%)
Allele distribution
G Carriers 94 (43.5%) 122 (56.5%) 2.079 (df = 1) 0.149
Non-carriers 11 (61.1%) 7 (38.9%)
A Carriers 49 (41.2%) 70 (51.8%) 1.337 (df = 1) 0.248
Non-carriers 56 (48.7%) 59 (51.3%)
rs9657182
Genotype PD patients Controls χ2 p value
GG 29 (27.6%) 23 (17.8%) 3.368 (df = 2) 0.186
AG 48 (45.7%) 70 (54.3%)
AA 28 (26.7%) 36 (24.9%)
Allele Patients
G 106 (50.5%) 116 (49.5%) 1.412 (df = 1) 0.235
A 104 (45%) 142 (55%)
Allele distribution
G Carriers 77 (45.3%) 93 (54.7%) 0.045 (df = 1) 0.832
Non-carriers 28 (43.8%) 36 (56.3%)
A Carriers 76 (41.83%) 106 (58.2%) 3.210 (df = 1) 0.073
Non-carriers 29 (55.8%) 23 (44.2%)
Table 3.Genotype frequency, allele distribution and age at onset of the PD patients.
rs7820268
Genotype EOPD LOPD χ2 p value OR (95% CI)
GG 34 (64.2%) 21 (42%) 6.352 (df = 2) 0.042 -
AG 13 (24.5%) 24 (48%)
AA 6 (11.3%) 5 (10%)
Allele distribution
G Carriers 47 (51.1%) 45 (48.9%) 0.047 (df = 1) 0.828 1.149 (0.327–4.031)
Non-carriers 6 (54.5%) 5 (45.5%)
A Carriers 19 (39.6%) 29 (60.4%) 5.073 (df = 1) 0.024 2.471 (1.117–5.467)
Non-carriers 34 (61.8%) 21 (38.2%)
rs9657182
Genotype EOPD LOPD χ2 p value OR (95% CI)
GG 19 (35.8%) 9 (18%) 4.795 (df = 2) 0.091 -
AG 23 (43.4%) 24 (48%)
AA 11 (20.8%) 17 (34%)
Allele distribution
G Carriers 42 (56%) 33 (44%) 2.280 (df = 1) 0.131 0.508 (0.210–1.232)
Non-carriers 11 (39.3%) 17 (60.7%)
A Carriers 34 (45.3%) 41 (54.7%) 4.141 (df = 1) 0.042 2.546 (1.020–6.351)
Non-carriers 19 (67.9%) 9 (32.1%)
EOPD, Early onset <60 years; LOPD, Late onset 60.

The genotype distribution rs9657182 was 29 GG, 48 GA and 28 AA in the PD group, and 23 GG, 70 GA and 36 AA in the control group. The allele frequency results did not show remarkable differences, with 50.5% G allele in the PD group vs 45% G allele in the controls, and 49.5% A allele in the PD group vs 55% A allele in the control group (Table 2). This SNP variant is likewise not associated with PD (genotype: p = 0.186, G allele: p = 0.832, A allele: p = 0.073), but it may influence the age at disease onset (genotype: p = 0.091, G allele: p = 0.131, A allele: p = 0.042) (Table 3). Similarly, the A allele seems to be a factor affecting the age at onset of PD (Table 3), as carrying the allele associates with LOPD (54.7% vs 45.3%), and non-carrying associates with EOPD (67.9% vs 32.1%) (OR = 2.546, 95% CI (1.020–6.351)), p = 0.042 for AA + AG vs GG).

4. Discussion

DA receptors, catechol-O-methyltransferase, monoamine oxidase B, NMDA receptors, adenosine A2A receptors, and cholinergic receptors are main targets of PD medication approved for clinical use. The drugs frequently cause serious side effects, become less effectual during treatment, and eventually lead to development of drug-resistant PD such as L-DOPA-resistant PD [52, 53]. Thus, a search for novel therapeutic targets is under extensive research. Intervention in the KP of TRP metabolism, adjacent biosystems, and the gut microbiota has been shed light on in search of novel biomarkers and drugs for neurodegenerative and psychiatric diseases [54, 55, 56, 57, 58, 59, 60, 61]. Glutamate and acetylcholine are main neurotransmitters responsible for cognition and behavior in which bioactive KYNs mediate neurotoxic, neuromodulatory, and immunological response in reaction to pathological insults [10, 54, 62].

The decreased concentrations of L-KYN and KYNA in the frontal cortex, putamen and SNpc, and elevated concentrations of 3-HK in the putamen and SNpc were observed in postmortem brain of PD patients [62, 63]. Clinical manifestations of dysregulation of kynurenine metabolism, including impairments in memory and learning (i.e., memory consolidation, poor planning, defects in set-shifting, impaired working memory, and executive dysfunction) are common in PD and correlate with a typical cognitive pattern due to prefrontal cortex dysfunction [64, 65, 66, 67, 68, 69, 70]. In addition to the central nervous system, peripheral samples showed abnormalities such as the increased activity of kynurenine aminotransferase (KAT) and elevated KYNA levels in red blood cells, which may be a protective response [71]. Natural product curcumin was reported to relieve pain and stress through the KYN metabolic pathway [72]. The KP is proposed to be a potential biomarker and target for treatment of Alzheimer’s diseases, schizophrenia, and depression [55, 72, 73, 74, 75, 76, 77]. Psychedelic psilocybin was reported to relieve depressive and anxiety symptoms of patients with terminal illness [78]. The action is considered at least partly through the KP. Furthermore, delivery of active agents to the brain through the blood-brain barrier is under extensive research [79]. Other in vitro and in vivo studies showed therapeutic opportunities through elevated levels of KYNA in PD [48, 80, 81].

However, in recent years growing body of evidence has accumulated on the importance of the KP in PD pathogenesis, only limited data are available on the genetic alterations of KP enzymes in PD yet. Until now, only two kynurenine enzymes, ACMSD and KMO genes polymorphisms have been investigated in PD [82, 83]. ACMSD play a key role in the KP, generating neuroactive metabolites which may play a crucial role in PD pathogenesis by inhibiting excitotoxicity and inflammation, therefore it could be a potential therapeutic target in PD [84]. Genome-wide association studies described that ACMSD polymorphisms are risk factors in PD [85, 86, 87], however its importance could not be confirmed in different populations [88, 89].

Previously, SNP analysis of KMO in PD and healthy controls were compared and a subgroup analysis was conducted according to age of onset. Four loci of SNPs did not reveal any significance between PD and healthy controls and between subgroups of age of onset [90].

In this study we compared the frequencies of three SNPs of IDO1 gene between PD patients and control individuals. Our results revealed that the investigated SNPs were not associated with PD, however, the rs7820268 and the rs9657182 SNPs showed significant associations with age of onset of the disease. The A allele of rs7820268 and rs9657182 SNPs associated with LOPD. It is well known that there are some genetic factors which show associations with age of onset in PD. Several mutations in PINK-1, parkin, LRRK2 and GBA genes are associated with EOPD. Our findings suggest that the A allele of these SNPs potentially delayed the development of PD [91, 92, 93, 94, 95].

The IDO1 gene is located on chromosome 8 spanning 14,900 nucleotides. It encodes the 403 amino acid IDO1 enzyme. IDO1 plays a crucial role in the production of immune and neuroactive KP metabolites, therefore its importance in immunomodulation and inflammatory processes is expected [96]. Previously have been described, that the minor allele of the rs7820268 SNP was shown to impair CD8+ regulatory T cell function [97]. The allele may lead to beneficial immunosuppressive effects in the rs7820268 SNP carriers in PD. The rs9657182 SNP is linked to moderate or severe IFN-α-induced depressive symptoms in Caucasian patients with chronic hepatitis C. The SNP is located in the linkage disequilibrium block encompassing 20 kilobases upstream of IDO1 as well as includes at least six additional polymorphisms that may contain binding sites for immunoregulatory proteins or may have epigenetic regulatory function [50]. Furthermore, the effects of endogenous metabolites and synthetic analogues on behavioral domains have been extensively studied in animal models [98, 99]. The downstream metabolite of the KP, kynurenic acid showed an antidepressant-like action in the animal model of depression [100]. Indoximod is a methylated TRP that inhibits IDO to maintain or elevate TRP levels important to T cell function. Interestingly, a case study reported that indoximod induced parkinsonism in a patient treated for metastatic breast cancer [101]. Recently, an IDO1 inhibitor (1-Methyltryptophan) was tested in murine model of PD, and the results showed improved locomotor activities, reduced level of neuroinflammatory and neuronal apoptotic markers. These findings suggest that the IDO1 inhibition could be a potential alternative in the pharmacotherapy of PD [102].

PD is a complex multifactorial disease, underlying mechanisms causing the pathological conditions of PD is not fully understood yet. In addition to the age, which is the most important risk factor for neurodegenerative disease, genetic, epigenetic, environmental and lifestyle are initiation factors which play a role in the pathogenesis of PD [16, 90, 103, 104, 105, 106, 107, 108, 109, 110, 111, 112, 113]. The preclinical studies, and diagnostic precision of PD are under extensive research, applying in vitro, in vivo, and in silico methodologies [114, 115, 116, 117, 118, 119]. Until now, 21 PARK genes have been described in human genome as causative factors of the disease, furthermore, genetic variants of 26 loci have been shown to be important risk modifiers for PD. Beside genetic factors, there are more evidence that epigenetic mechanisms, like DNA methylation, histone modifications and dysregulation of non-coding RNAs (e.g., long non-coding RNA and microRNA) play a role in pathogenesis of PD [109, 120]. Several studies described that the gene expression of different genes is altered in PD, which may help predict the PD progression, and identify new targets for therapeutic intervention [121, 122, 123]. Moreover, it is evident that the KP are involved in the pathogenesis of PD. Our result, in line with data of others’, strengthen the involvement of IDO1 in the pathogenesis of PD. Furthermore, the influence of SNPs on the non-motor symptoms of PD is of particular interest. The gene variants of alpha-synuclein SNCA rs11931074 and glucosylceramidase beta 1 rs708606 have been reported to correlate with hyposomnia in PD [124, 125]. The direct involvement of KP metabolism in the non-motor symptoms of PD is to be explored.

This study had several limitations including its relatively small sample size, the small number of SNP loci, heterogeneity of the study subjects, and the collection of blood samples at a single center. Furthermore, enzyme activities of IDO1, IDO2, and TDO were not measured. Thus, it was not known if IDO2 and/or TDO compensated the polymorphic variants of IDO1. Prospective studies in larger patient number, with the larger number of SNP loci, and data with IDO2 and TDO activities are needed to validate our findings.

5. Conclusions

This study explored the potential role of IDO1 gene polymorphisms in PD. None of three IDO1 SNPs investigated in this study were significantly associated with PD; however, two IDO1 SNPs showed correlated with the age onset of PD, suggesting that the gene polymorphisms may not play a direct role in pathogenesis of PD, but may influence the disease onset probably as secondary risk factors. The further investigation is expected in search of roles of gene polymorphism in risk, onset, prognosis, progression of PD including SNPs, structural variants, and the disease-related pathways.

Abbreviations

AMPA, 2-amino-3-carboxymuconate-semialdehyde decarboxylase; AMPA, alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid; CSF, cerebrospinal fluid; DA, dopamine; 3-HAA, 3-hydroxyanthranillic acid; 3-HK, 3-hydroxykynurenine; IDO, indoleamine 2,3-dioxygenase; IFN, interferon; KAT, kynurenine aminotransferases; KMO, kynurenine 3-monooxygenase; KP, kynurenine pathway; KYNA, kynurenic acid; NMDA, N-methyl-D-aspartate; PD, Parkinson’s disease; QUIN, quinolinic acid; ROS, reactive oxygen species; SNP, single nucleotide polymorphisms; TDO, tryptopan 2,3-dioxygenase; TRP, tryptophan.

Author Contributions

NT designed the research study. NT performed the research. KB analyzed the data. NT, RMT, KM, ZS, FS, KB, MT, PK and LV wrote the manuscript. All authors contributed to editorial changes in the manuscript. All authors read and approved the final manuscript.

Ethics Approval and Consent to Participate

All patients gave their informed consent in accordance with the Declaration of Helsinki, and the study was approved by the Medical Research Council Scientific and Research Ethics Committee (47066-3/2013/EKU (556/2013)).

Acknowledgment

The authors are sincerely grateful to Dr. Michael D. Lovelace and his co-workers for critical reading and their kind help.

Funding

This research was funded by the Economic Development and Innovation Operational Programme [GINOP 2.3.2-15-2016-00034; GINOP 2.3.2-15-2016-00048], National Research, Development and Innovation Office [NKFIH-1279-2/2020 TKP 2020], TUDFO/47138-1/2019-ITM, National Scientific Research Fund [OTKA138125] and ELKH-SZTE. Project no. TKP2021-EGA-32 has been implemented with the support provided by the Ministry of Innovation and Technology of Hungary from the National Research, Development and Innovation Fund, financed under the TKP2021-EGA funding scheme.section is required for all papers.

Conflict of Interest

Given his role as Guest Editor, Masaru Tanaka had no involvement in the peer-review of this article and has no access to information regarding its peer-review. Full responsibility for the editorial process for this article was delegated to Graham Pawelec. The authors declare no conflict of interest.

Publisher’s Note: IMR Press stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

References
[1]
Pringsheim T, Jette N, Frolkis A, Steeves TDL. The prevalence of Parkinson’s disease: a systematic review and meta-analysis. Movement Disorders. 2014; 29: 1583–1590.
[2]
Wu T, Hallett M, Chan P. Motor automaticity in Parkinson’s disease. Neurobiology of Disease. 2015; 82: 226–234.
[3]
Tanaka M, Vécsei L. Editorial of Special Issue ‘Dissecting Neurological and Neuropsychiatric Diseases: Neurodegeneration and Neuroprotection’. International Journal of Molecular Sciences. 2022; 23: 6991.
[4]
El-Kattan MM, Rashed LA, Shazly SR, Ismail RS. Relation of serum level of tumor necrosis factor-alpha to cognitive functions in patients with Parkinson’s disease. The Egyptian Journal of Neurology, Psychiatry and Neurosurgery. 2022; 58: 25.
[5]
Tanaka M, Vécsei L. Editorial of Special Issue “Crosstalk between Depression, Anxiety, and Dementia: Comorbidity in Behavioral Neurology and Neuropsychiatry”. Biomedicines. 2021; 9: 517.
[6]
Tanaka M. Crosstalk between Depression, Anxiety, and Dementia: Comorbidity in Behavioral Neurology and Neuropsychiatry (pp. 1–266). 1st edn. MDPI: Basel, Switzerland. 2022.
[7]
Byeon H. Development of a Stacking-Based Ensemble Machine Learning for Detection of Depression in Parkinson’s Disease: Preliminary Research. Biology Life Sciences Forum. 2021; 9: 5.
[8]
Cao X, Yang F, Zheng J, Wang X, Huang Q. Aberrant Structure MRI in Parkinson’s Disease and Comorbidity with Depression Based on Multinomial Tensor Regression Analysis. Journal of Personalized Medicine. 2022; 12: 89.
[9]
Dickson DW, Fujishiro H, Orr C, DelleDonne A, Josephs KA, Frigerio R, et al. Neuropathology of non-motor features of Parkinson disease. Parkinsonism and Related Disorders. 2009; 15: S1–S5.
[10]
Tanaka M, Bohár Z, Vécsei L. Are Kynurenines Accomplices or Principal Villains in Dementia? Maintenance of Kynurenine Metabolism. Molecules. 2020; 25: 564.
[11]
Ellena G, Battaglia S, Làdavas E. The spatial effect of fearful faces in the autonomic response. Experimental Brain Research. 2020; 238: 2009–2018.
[12]
Borgomaneri S, Vitale F, Battaglia S, Avenanti A. Early Right Motor Cortex Response to Happy and Fearful Facial Expressions: A TMS Motor-Evoked Potential Study. Brain Sciences. 2021; 11: 1203.
[13]
Candini M, Battaglia S, Benassi M, di Pellegrino G, Frassinetti F. The physiological correlates of interpersonal space. Scientific Reports. 2021; 11: 2611.
[14]
Battaglia S, Thayer JF. Functional interplay between central and autonomic nervous systems in human fear conditioning. Trends in Neurosciences. 2022; 45: 504–506.
[15]
Battaglia S, Orsolini S, Borgomaneri S, Barbieri R, Diciotti S, di Pellegrino G. Characterizing cardiac autonomic dynamics of fear learning in humans. Psychophysiology. 2022; e14122.
[16]
Tanaka M, Toldi J, Vecsei L. Exploring the Etiological Links behind Neurodegenerative Diseases: Inflammatory Cytokines and Bioactive Kynurenines. International Journal of Molecular Sciences. 2020; 21: 2431.
[17]
Schirru M, Véronneau-Veilleux F, Nekka F, Ursino M. Phasic Dopamine Changes and Hebbian Mechanisms during Probabilistic Reversal Learning in Striatal Circuits: A Computational Study. International Journal of Molecular Sciences. 2022; 23: 3452.
[18]
Tanaka M, Spekker E, Szabó Á, Polyák H, Vécsei L. Modelling the neurodevelopmental pathogenesis in neuropsychiatric disorders. Bioactive kynurenines and their analogues as neuroprotective agents-in celebration of 80th birthday of Professor Peter Riederer. Journal of Neural Transmission. 2022; 129: 627–642.
[19]
Caudle WM, Zhang J. Glutamate, excitotoxicity, and programmed cell death in parkinson disease. Experimental Neurology. 2009; 220: 230–233.
[20]
Reale M, Iarlori C, Thomas A, Gambi D, Perfetti B, Di Nicola M, et al. Peripheral cytokines profile in Parkinson’s disease. Brain, Behavior, and Immunity. 2009; 23: 55–63.
[21]
Blandini F. An update on the potential role of excitotoxicity in the pathogenesis of Parkinson’s disease. Functional Neurology. 2010; 25: 65–71.
[22]
Torok N, Majlath Z, Szalardy L, Vecsei L. Investigational alpha-synuclein aggregation inhibitors: hope for Parkinson’s disease. Expert Opinion on Investigational Drugs. 2016; 25: 1281–1294.
[23]
Spatola M, Wider C. Genetics of Parkinson’s disease: the yield. Parkinsonism and Related Disorders. 2014; 20: S35–S38.
[24]
Török N, Majláth Z, Fülöp F, Toldi J, Vécsei L. Brain Aging and Disorders of the Central Nervous System: Kynurenines and Drug Metabolism. Current Drug Metabolism. 2016; 17: 412–429.
[25]
Tanaka M, Szabó Á, Spekker E, Polyák H, Tóth F, Vécsei L. Mitochondrial Impairment: A Common Motif in Neuropsychiatric Presentation? The Link to the Tryptophan-Kynurenine Metabolic System. Cells. 2022; 11: 2607.
[26]
Tan LY, Tang, KH, Lim LYY, Ong JX, Park H, Jung S. α-Synuclein at the Presynaptic Axon Terminal as a Double-Edged Sword. Biomolecules. 2022; 12: 507.
[27]
Hsu YL, Hung HS, Tsai CW, Liu SP, Chiang YT, Kuo YH, et al. Peiminine Reduces ARTS-Mediated Degradation of XIAP by Modulating the PINK1/Parkin Pathway to Ameliorate 6-Hydroxydopamine Toxicity and α-Synuclein Accumulation in Parkinson’s Disease Models In Vivo and In Vitro. International Journal of Molecular Sciences. 2021; 22: 10240.
[28]
Hunt C, Macedo e Cordeiro T, Suchting R, de Dios C, Cuellar Leal VA, Soares JC, et al. Effect of immune activation on the kynurenine pathway and depression symptoms - a systematic review and meta-analysis. Neuroscience and Biobehavioral Reviews. 2020; 118: 514–523.
[29]
Cao B, Chen Y, Ren Z, Pan Z, McIntyre RS, Wang D. Dysregulation of kynurenine pathway and potential dynamic changes of kynurenine in schizophrenia: a systematic review and meta-analysis. Neuroscience and Biobehavioral Reviews. 2021; 123: 203–214.
[30]
Jovanovic F, Candido KD, Knezevic NN. The Role of the Kynurenine Signaling Pathway in Different Chronic Pain Conditions and Potential Use of Therapeutic Agents. International Journal of Molecular Sciences. 2020; 21: 6045.
[31]
Carrillo-Mora P, Pérez-De la Cruz V, Estrada-Cortés B, Toussaint-González P, Martínez-Cortéz JA, Rodríguez-Barragán M, et al. Serum Kynurenines Correlate with Depressive Symptoms and Disability in Poststroke Patients: a Cross-sectional Study. Neurorehabilitation and Neural Repair. 2020; 34: 936–944.
[32]
Tanaka M, Vécsei L. Monitoring the kynurenine system: Concentrations, ratios or what else? Advances in Clinical and Experimental Medicine. 2021; 30: 775–778.
[33]
Spekker E, Tanaka M, Szabó Á, Vécsei L. Neurogenic Inflammation: The Participant in Migraine and Recent Advancements in Translational Research. Biomedicines. 2021; 10: 76.
[34]
Tanaka M, Török N, Tóth F, Szabó Á, Vécsei L. Co-Players in Chronic Pain: Neuroinflammation and the Tryptophan-Kynurenine Metabolic Pathway. Biomedicines. 2021; 9: 897.
[35]
Simonato M, Dall’Acqua S, Zilli C, Sut S, Tenconi R, Gallo N, et al. Tryptophan Metabolites, Cytokines, and Fatty Acid Binding Protein 2 in Myalgic Encephalomyelitis/Chronic Fatigue Syndrome. Biomedicines. 2021; 9: 1724.
[36]
Jamshed L, Debnath A, Jamshed S, Wish JV, Raine JC, Tomy GT, et al. An Emerging Cross-Species Marker for Organismal Health: Tryptophan-Kynurenine Pathway. International Journal of Molecular Sciences. 2022; 23: 6300.
[37]
Widner B, Leblhuber F, Fuchs D. Increased neopterin production and tryptophan degradation in advanced Parkinson’s disease. Journal of Neural Transmission. 2002; 109: 181–189.
[38]
Ramírez Ortega D, Ugalde Muñiz PE, Blanco Ayala T, Vázquez Cervantes GI, Lugo Huitrón R, Pineda B, et al. On the Antioxidant Properties of L-Kynurenine: An Efficient ROS Scavenger and Enhancer of Rat Brain Antioxidant Defense. Antioxidants. 2022; 11: 31.
[39]
Romero R, Chaemsaithong P, Yeo L, Docheva N, Chaiyasit N. Fetal and Maternal Responses to Intraamniotic Infection. Fetal and Neonatal Physiology. 2017; 208: 144–159.e12.
[40]
Majláth Z, Török N, Toldi J, Vécsei L. Memantine and Kynurenic Acid: Current Neuropharmacological Aspects. Current Neuropharmacology. 2016; 14: 200–209.
[41]
Tuboly G, Tar L, Bohar Z, Safrany-Fark A, Petrovszki Z, Kekesi G, et al. The inimitable kynurenic acid: the roles of different ionotropic receptors in the action of kynurenic acid at a spinal level. Brain Research Bulletin. 2015; 112: 52–60.
[42]
Prescott C, Weeks AM, Staley KJ, Partin KM. Kynurenic acid has a dual action on AMPA receptor responses. Neuroscience Letters. 2006; 402: 108–112.
[43]
Rózsa E, Robotka H, Vécsei L, Toldi J. The Janus-face kynurenic acid. Journal of Neural Transmission. 2008; 115: 1087–1091.
[44]
Martos D, Tuka B, Tanaka M, Vécsei L, Telegdy G. Memory Enhancement with Kynurenic Acid and Its Mechanisms in Neurotransmission. Biomedicines. 2022; 10: 849.
[45]
Stone TW. Does kynurenic acid act on nicotinic receptors? an assessment of the evidence. Journal of Neurochemistry. 2020; 152: 627–649.
[46]
Tomaszewska E, Muszynski S, Kuc D, Dobrowolski P, Lamorski K, Smolińska K, et al. Chronic dietary supplementation with kynurenic acid, a neuroactive metabolite of tryptophan, decreased body weight without negative influence on densitometry and mandibular bone biomechanical endurance in young rats. PLoS ONE. 2019; 14: e0226205.
[47]
Mándi Y, Vécsei L. The kynurenine system and immunoregulation. Journal of Neural Transmission. 2012; 119: 197–209.
[48]
Lee DY, Lee K, Lee HJ, Noh YH, Kim DH, Lee JY, et al. Kynurenic acid attenuates MPP+-induced dopaminergic neuronal cell death via a Bax-mediated mitochondrial pathway. European Journal of Cell Biology. 2008; 87: 389–397.
[49]
GREGOIRE L, RASSOULPOUR A, GUIDETTI P, SAMADI P, BEDARD P, IZZO E, et al. Prolonged kynurenine 3-hydroxylase inhibition reduces development of levodopa-induced dyskinesias in parkinsonian monkeys. Behavioural Brain Research. 2008; 186: 161–167.
[50]
Smith AK, Simon JS, Gustafson EL, Noviello S, Cubells JF, Epstein MP, et al. Association of a polymorphism in the indoleamine-2,3-dioxygenase gene and interferon-alpha-induced depression in patients with chronic hepatitis C. Molecular Psychiatry. 2012; 17: 781–789.
[51]
Miller SA, Dykes DD, Polesky HF. A simple salting out procedure for extracting DNA from human nucleated cells. Nucleic Acids Research. 1988; 16: 1215–1215.
[52]
Dézsi L, Vécsei L. Clinical implications of irregular ADMET properties with levodopa and other antiparkinson’s drugs. Expert Opinion on Drug Metabolism and Toxicology. 2014; 10: 409–424.
[53]
Calabresi P, Ghiglieri V, Mazzocchetti P, Corbelli I, Picconi B. Levodopa-induced plasticity: a double-edged sword in Parkinson’s disease? Philosophical Transactions of the Royal Society B: Biological Sciences. 2015; 370: 20140184.
[54]
Tanaka M, Török N, Vécsei L. Novel Pharmaceutical Approaches in Dementia. NeuroPsychopharmacotherapy. 2021; 24: 1–18.
[55]
Tanaka M, Török N, Vécsei L. Are 5-HT_1 receptor agonists effective anti-migraine drugs? Expert Opinion on Pharmacotherapy. 2021; 22: 1221–1225.
[56]
Török N, Tanaka M, Vécsei L. Searching for Peripheral Biomarkers in Neurodegenerative Diseases: The Tryptophan-Kynurenine Metabolic Pathway. International Journal of Molecular Sciences. 2020; 21: 9338.
[57]
Ciapała K, Mika J, Rojewska E. The Kynurenine Pathway as a Potential Target for Neuropathic Pain Therapy Design: From Basic Research to Clinical Perspectives. International Journal of Molecular Sciences. 2021; 22: 11055.
[58]
Fila M, Chojnacki J, Pawlowska E, Szczepanska J, Chojnacki C, Blasiak J. Kynurenine Pathway of Tryptophan Metabolism in Migraine and Functional Gastrointestinal Disorders. International Journal of Molecular Sciences. 2021; 22: 10134.
[59]
González-Sanmiguel J, Schuh CMAP, Muñoz-Montesino C, Contreras-Kallens P, Aguayo LG, Aguayo S. Complex Interaction between Resident Microbiota and Misfolded Proteins: Role in Neuroinflammation and Neurodegeneration. Cells. 2020; 9: 2476.
[60]
Sun P, Su L, Zhu H, Li X, Guo Y, Du X, et al. Gut Microbiota Regulation and Their Implication in the Development of Neurodegenerative Disease. Microorganisms. 2021; 9: 2281.
[61]
Di Gregorio F, La Porta F, Petrone V, Battaglia S, Orlandi S, Ippolito G, et al. Accuracy of EEG Biomarkers in the Detection of Clinical Outcome in Disorders of Consciousness after Severe Acquired Brain Injury: Preliminary Results of a Pilot Study Using a Machine Learning Approach. Biomedicines. 2022; 10: 1897.
[62]
Stone TW, Darlington LG. The kynurenine pathway as a therapeutic target in cognitive and neurodegenerative disorders. British Journal of Pharmacology. 2013; 169: 1211–1227.
[63]
Ogawa T, Matson WR, Beal MF, Myers RH, Bird ED, Milbury P, et al. Kynurenine pathway abnormalities in Parkinson’s disease. Neurology. 1992; 42: 1702–1702.
[64]
Battaglia S, Garofalo S, di Pellegrino G, Starita F. Revaluing the Role of vmPFC in the Acquisition of Pavlovian Threat Conditioning in Humans. The Journal of Neuroscience. 2020; 40: 8491–8500.
[65]
Battaglia S, Harrison BJ, Fullana MA. Does the human ventromedial prefrontal cortex support fear learning, fear extinction or both? a commentary on subregional contributions. Molecular Psychiatry. 2022; 27: 784–786.
[66]
Battaglia S, Fabius JH, Moravkova K, Fracasso A, Borgomaneri S. The Neurobiological Correlates of Gaze Perception in Healthy Individuals and Neurologic Patients. Biomedicines. 2022; 10: 627.
[67]
Garofalo S, Battaglia S, Starita F, di Pellegrino G. Modulation of cue-guided choices by transcranial direct current stimulation. Cortex. 2021; 137: 124–137.
[68]
Garofalo S, Battaglia, Di Fazio C, Nazzi C, Fracasso A, Borgomaneri S. The Influence of Vicarious Fear-Learning in “Infecting” Reactive Action Inhibition. Frontierrs in Behavioral Neurosciences. 2022; 16: 946263.
[69]
Sellitto M, Terenzi D, Starita F, di Pellegrino G, Battaglia S. The Cost of Imagined Actions in a Reward-Valuation Task. Brain Sciences. 2022; 12: 582.
[70]
Garofalo S, Timmermann C, Battaglia S, Maier ME, di Pellegrino G. Mediofrontal Negativity Signals Unexpected Timing of Salient Outcomes. Journal of Cognitive Neuroscience. 2017; 29: 718–727.
[71]
Hartai Z, Klivenyi P, Janaky T, Penke B, Dux L, Vecsei L. Kynurenine metabolism in plasma and in red blood cells in Parkinson’s disease. Journal of the Neurological Sciences. 2005; 239: 31–35.
[72]
Zhang Y, Li L, Zhang J. Curcumin in antidepressant treatments: an overview of potential mechanisms, pre‐clinical/clinical trials and ongoing challenges. Basic and Clinical Pharmacology and Toxicology. 2020; 127: 243–253.
[73]
Koola M, Jafarnejad S, Looney S, Praharaj S, Pillai A, Ahmed A, et al. Meta-Analyses of Randomized Controlled Trials and Potential Novel Combination Treatments in Schizophrenia. Biological Psychiatry. 2020; 87: S306.
[74]
Koola MM. Galantamine-Memantine combination in the treatment of Alzheimer’s disease and beyond. Psychiatry Research. 2020; 293: 113409.
[75]
Hestad K, Alexander J, Rootwelt H, Aaseth JO. The Role of Tryptophan Dysmetabolism and Quinolinic Acid in Depressive and Neurodegenerative Diseases. Biomolecules. 2022; 12: 998.
[76]
Kucukkarapinar M, Yay-Pence A, Yildiz Y, Buyukkoruk M, Yaz-Aydin G, Deveci-Bulut TS, et al. Psychological outcomes of COVID-19 survivors at sixth months after diagnose: the role of kynurenine pathway metabolites in depression, anxiety, and stress. Journal of Neural Transmission. 2022; 129: 1077–1089.
[77]
Erabi H, Okada G, Shibasaki C, Setoyama D, Kang D, Takamura M, et al. Kynurenic acid is a potential overlapped biomarker between diagnosis and treatment response for depression from metabolome analysis. Scientific Reports. 2020; 10: 16822.
[78]
Vargas AS, Luis A, Barroso M, Gallardo E, Pereira L. Psilocybin as a New Approach to Treat Depression and Anxiety in the Context of Life-Threatening Diseases-A Systematic Review and Meta-Analysis of Clinical Trials. Biomedicines. 2020; 8: 331.
[79]
Rog J, Blazewicz A, Juchnowicz D, Ludwiczuk A, Stelmach E, Kozioł M, et al. The Role of GPR120 Receptor in Essential Fatty Acids Metabolism in Schizophrenia. Biomedicines. 2020; 8: 243.
[80]
Butler EG, Bourke DW, Finkelstein DI, Horne MK. The effects of reversible inactivation of the subthalamo-pallidal pathway on the behaviour of naive and hemiparkinsonian monkeys. Journal of Clinical Neuroscience. 1997; 4: 218–227.
[81]
Miranda AF, Boegman RJ, Beninger RJ, Jhamandas K. Protection against quinolinic acid-mediated excitotoxicity in nigrostriatal dopaminergic neurons by endogenous kynurenic acid. Neuroscience. 1997; 78: 967–975.
[82]
Venkatesan D, Iyer M, Narayanasamy A, Siva K, Vellingiri B. Kynurenine pathway in Parkinson’s disease—an update. ENeurologicalSci. 2020; 21: 100270.
[83]
Lim CK, Fernández-Gomez FJ, Braidy N, Estrada C, Costa C, Costa S, et al. Involvement of the kynurenine pathway in the pathogenesis of Parkinson’s disease. Progress in Neurobiology. 2017; 155: 76–95.
[84]
Thirtamara-Rajamani K, Li P, Escobar Galvis ML, Labrie V, Brundin P, Brundin L. Is the Enzyme ACMSD a Novel Therapeutic Target in Parkinson’s Disease? Journal of Parkinson’s Disease. 2017; 7: 577–587.
[85]
Lill CM, Roehr JT, McQueen MB, Kavvoura FK, Bagade S, Schjeide BM, et al. Comprehensive research synopsis and systematic meta-analyses in Parkinson’s disease genetics: The PDGene database. PLoS Genetics. 2012; 8: e1002548.
[86]
Nalls MA, Plagnol V, Hernandez DG, Sharma M, Sheerin U, Saad M, et al. Imputation of sequence variants for identification of genetic risks for Parkinson’s disease: a meta-analysis of genome-wide association studies. The Lancet. 2011; 377: 641–649.
[87]
Bandrés-Ciga S, Price TR, Barrero FJ, Escamilla-Sevilla F, Pelegrina J, Arepalli S, et al. Genome-wide assessment of Parkinson’s disease in a Southern Spanish population. Neurobiology of Aging. 2016; 45: 213.e3–213.e9.
[88]
Wang L, Li N, Lu Z, Li J, Peng J, Duan L, et al. Association of three candidate genetic variants in ACMSD/TMEM163, GPNMB and BCKDK /STX1B with sporadic Parkinson’s disease in Han Chinese. Neuroscience Letters. 2019; 703: 45–48.
[89]
Chang KH, Chen CM, Chen YC, Fung HC, Wu YR. Polymorphisms of ACMSD-TMEM163, MCCC1, and BCKDK-STX1B Are Not Associated with Parkinson’s Disease in Taiwan. Parkinson’s Disease. 2019; 2019: 3489638.
[90]
Török N, Török R, Szolnoki Z, Somogyvári F, Klivényi P, Vécsei L. The Genetic Link between Parkinson’s Disease and the Kynurenine Pathway is still Missing. Parkinson’s Disease. 2015; 2015: 1–7.
[91]
Török R, Zádori D, Török N, Csility É, Vécsei L, Klivényi P. An assessment of the frequency of mutations in the GBA and VPS35 genes in Hungarian patients with sporadic Parkinson’s disease. Neuroscience Letters. 2016; 610: 135–138.
[92]
Lucking CB, Durr A, Bonifati V, Vaughan J, De Michele G, Gasser T, et al. Association between early-onset Parkinson’s disease and mutations in the parkin gene. The New England Journal of Medicine. 2000; 342: 1560–1567.
[93]
Bonifati V, Rohe CF, Breedveld GJ, Fabrizio E, De Mari M, Tassorelli C, et al. Early-onset parkinsonism associated with PINK1 mutations: Frequency, genotypes, and phenotypes. Neurology. 2005; 65: 87–95.
[94]
Healy DG, Falchi M, O’sullivan SS, Bonifati V, Durr A, Bressman S, et al. Phenotype, genotype, and worldwide genetic penetrance of LRRK2-associated Parkinson’s disease: a case-control study. The Lancet Neurology. 2008; 7: 583–590.
[95]
Chen Y, Gu X, Ou R, Zhang L, Hou Y, Liu K, et al. Evaluating the Role of SNCA, LRRK2, and GBA in Chinese Patients With Early-Onset Parkinson’s Disease. Movement Disordders. 2020; 35: 2046–2055.
[96]
Boros FA, Vecsei L. Immunomodulatory Effects of Genetic Alterations Affecting the Kynurenine Pathway. Frontiers in Immunology. 2019; 10: 2570.
[97]
Tardito S, Negrini S, Conteduca G, Ferrera F, Parodi A, Battaglia F, et al. Indoleamine 2,3 dioxygenase gene polymorphisms correlate with CD8+ Treg impairment in systemic sclerosis. Human Immunology. 2013; 74: 166–169.
[98]
Telegdy G, Adamik A, Tanaka M, Schally AV. Effects of the LHRH antagonist Cetrorelix on affective and cognitive functions in rats. Regulatory Peptides. 2010; 159: 142–147.
[99]
Telegdy G, Tanaka M, Schally AV. Effects of the growth hormone-releasing hormone (GH-RH) antagonist on brain functions in mice. Behavioural Brain Research. 2011; 224: 155–158.
[100]
Tanaka M, Bohár Z, Martos D, Telegdy G, Vécsei L. Antidepressant-like effects of kynurenic acid in a modified forced swim test. Pharmacological Reports. 2020; 72: 449–455.
[101]
Floyd M, El Osta B, Tang S. First Report of Parkinsonism Associated with Indoximod, an Immune-Modulating Agent. Journal of Global Oncology. 2018; 4: 1–2.
[102]
Sodhi RK, Bansal Y, Singh R, Saroj P, Bhandari R, Kumar B, et al. IDO-1 inhibition protects against neuroinflammation, oxidative stress and mitochondrial dysfunction in 6-OHDA induced murine model of Parkinson’s disease. NeuroToxicology. 2021; 84: 184–197.
[103]
Aasly JO. Long-Term Outcomes of Genetic Parkinson’s Disease. Journal of Movement Disorders. 2020; 13: 81–96.
[104]
Marki S, Goblos A, Szlavicz E, Török N, Balicza P, Bereznai B, et al. The rs13388259 Intergenic Polymorphism in the Genomic Context of the BCYRN1 Gene Is Associated with Parkinson’s Disease in the Hungarian Population. Parkinson’s Disease. 2018; 2018: 9351598.
[105]
Nalls MA, Pankratz N, Lill CM, Do CB, Hernandez DG, Saad M, et al. Large-scale meta-analysis of genome-wide association data identifies six new risk loci for Parkinson’s disease. Nature Genetics. 2014; 46: 989–993.
[106]
Hall A, Bandres-Ciga S, Diez-Fairen M, Quinn JP, Billingsley KJ. Genetic Risk Profiling in Parkinson’s Disease and Utilizing Genetics to Gain Insight into Disease-Related Biological Pathways. International Journal of Molecular Sciences. 2020; 21: 7332.
[107]
Navarro-Sanchez L, Agueda-Gomez B, Aparicio S, Perez-Tur J. Epigenetic Study in Parkinson’s Disease: A Pilot Analysis of DNA Methylation in Candidate Genes in Brain. Cells. 2018; 7: 150.
[108]
Brito LM, Ribeiro-Dos-Santos A, Vidal AF, de Araujo GS. Differential Expression and miRNA-Gene Interactions in Early and Late Mild Cognitive Impairment. Biology. 2020; 9: 251.
[109]
Catanesi M, d’Angelo M, Tupone MG, Benedetti E, Giordano A, Castelli V, et al. MicroRNAs Dysregulation and Mitochondrial Dysfunction in Neurodegenerative Diseases. International Journal of Molecular Sciences. 2020; 21: 5986.
[110]
Sini P, Dang TBC, Fais M, Galioto M, Padedda BM, Lugliè A, et al. Cyanobacteria, Cyanotoxins, and Neurodegenerative Diseases: Dangerous Liaisons. International Journal of Molecular Sciences. 2021; 22: 8726.
[111]
Hepsomali P, Coxon C. Inflammation and diet: Focus on mental and cognitive health. Advances in Clinical and Experimental Medicine. 2022. (in press)
[112]
Chen C. Recent advances in the study of the comorbidity of depressive and anxiety disorders. Advances in Clinical and Experimental Medicine. 2022; 31: 355–358.
[113]
Gładka A, Zatoński T, Rymaszewska J. Association between the long-term exposure to air pollution and depression. Advances in Clinical and Experimental Medicine. 2022. (in press)
[114]
Datki Z, Sinka R. Translational biomedicine-oriented exploratory research on bioactive rotifer-specific biopolymers. Advances in Clinical and Experimental Medicine. 2022. (in press)
[115]
Mendes-Pinheiro B, Soares-Cunha C, Marote A, Loureiro-Campos E, Campos J, Barata-Antunes S, et al. Unilateral Intrastriatal 6-Hydroxydopamine Lesion in Mice: A Closer Look into Non-Motor Phenotype and Glial Response. International Journal of Molecular Sciences. 2021; 22: 11530.
[116]
Duarte-Jurado AP, Gopar-Cuevas Y, Saucedo-Cardenas O, Loera-Arias MJ, Montes-de-Oca-Luna R, Garcia-Garcia A, et al. Antioxidant Therapeutics in Parkinson’s Disease: Current Challenges and Opportunities. Antioxidants. 2021; 10: 453.
[117]
Pfaff AL, Bubb VJ, Quinn JP, Koks S. An Increased Burden of Highly Active Retrotransposition Competent L1s Is Associated with Parkinson’s Disease Risk and Progression in the PPMI Cohort. International Journal of Molecular Sciences. 2020; 21: 6562.
[118]
Hall A, Bandres-Ciga S, Diez-Fairen M, Quinn JP, Billingsley KJ. Genetic Risk Profiling in Parkinson’s Disease and Utilizing Genetics to Gain Insight into Disease-Related Biological Pathways. International Journal of Molecular Sciences. 2020; 21: 7332.
[119]
Lam S, Hartmann N, Benfeitas R, Zhang C, Arif M, Turkez H, et al. Systems Analysis Reveals Ageing-Related Perturbations in Retinoids and Sex Hormones in Alzheimer’s and Parkinson’s Diseases. Biomedicines. 2021; 9: 1310.
[120]
Boros FA, Maszlag-Török R, Vécsei L, Klivényi P. Increased level of NEAT1 long non-coding RNA is detectable in peripheral blood cells of patients with Parkinson’s disease. Brain Research. 2020; 1730: 146672.
[121]
Pinho R, Guedes LC, Soreq L, Lobo PP, Mestre T, Coelho M, et al. Gene Expression Differences in Peripheral Blood of Parkinson’s Disease Patients with Distinct Progression Profiles. PLoS ONE. 2016; 11: e0157852.
[122]
Henderson AR, Wang Q, Meechoovet B, Siniard AL, Naymik M, De Both M, et al. DNA Methylation and Expression Profiles of Whole Blood in Parkinson’s Disease. Frontiers in Genetics. 2021; 12: 640266.
[123]
Maszlag-Török R, Boros FA, Vécsei L, Klivényi P. Gene variants and expression changes of SIRT1 and SIRT6 in peripheral blood are associated with Parkinson’s disease. Scientific Reports. 2021; 11: 10677.
[124]
Chen W, Kang WY, Chen S, Wang Y, Xiao Q, Wang G, et al. Hyposmia correlates with SNCA variant and non-motor symptoms in Chinese patients with Parkinson’s disease. Parkinsonism Related Disorders. 2015; 21: 610–614.
[125]
Wilke MVMB, Dornelles AD, Schuh AS, Vairo FP, Basgalupp SP, Siebert M, et al. Evaluation of the frequency of non-motor symptoms of Parkinson’s disease in adult patients with Gaucher disease type 1. Orphanet Journal of Rare Diseases. 2019; 14: 103.
Share
Back to top