Next Article in Journal
The Relationship between Retained Primitive Reflexes and Hemispheric Connectivity in Autism Spectrum Disorders
Next Article in Special Issue
Neurological Insights into Sleep Disorders in Parkinson’s Disease
Previous Article in Journal
Involvement of Nrf2 Activation and NF-kB Pathway Inhibition in the Antioxidant and Anti-Inflammatory Effects of Hesperetin in Activated BV-2 Microglial Cells
Previous Article in Special Issue
Parkinson’s Disease: A Narrative Review on Potential Molecular Mechanisms of Sleep Disturbances, REM Behavior Disorder, and Melatonin
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

The Genetic Basis of Probable REM Sleep Behavior Disorder in Parkinson’s Disease

1
Observatorio de Salud Pública, Vicerrectorado de Investigación e Innovación Académica, Pontificia Universidad Católica Argentina (UCA), Consejo Nacional de Investigaciones Científicas y Técnicas (UCA-CONICET), Buenos Aires C1107AAZ, Argentina
2
Centro de Investigaciones en Psicología y Psicopedagogía (CIPP), Facultad de Psicología y Psicopedagogía, Pontificia Universidad Católica Argentina (UCA), Buenos Aires C1107AFB, Argentina
3
Departamento de Fisiología, Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires C1053ABH, Argentina
4
Centro de Altos Estudios en Ciencias Humanas y de la Salud, Universidad Abierta Interamericana, Consejo Nacional de Investigaciones Científicas y Técnicas, CAECIHS.UAI-CONICET, Buenos Aires C1270AAH, Argentina
5
Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Santiago 7500912, Chile
*
Author to whom correspondence should be addressed.
Brain Sci. 2023, 13(8), 1146; https://doi.org/10.3390/brainsci13081146
Submission received: 27 June 2023 / Revised: 25 July 2023 / Accepted: 27 July 2023 / Published: 30 July 2023
(This article belongs to the Special Issue Sleep Disorders in Parkinson’s Disease)

Abstract

:
Patients with Parkinson’s Disease (PD) experience REM sleep behavior disorder (RBD) more frequently than healthy controls. RBD is associated with torpid disease evolution. To test the hypothesis that differential genetic signatures might contribute to the torpid disease evolution in PD patients with RBD we compared the rate of genetic mutations in PD patients with or without probable RBD. Patients with a clinical diagnosis of PD in the Parkinson’s Progression Markers Initiative (PPMI) database entered the study. We excluded those with missing data, dementia, psychiatric conditions, or a diagnosis change over the first five years from the initial PD diagnosis. Probable RBD (pRBD) was confirmed by a REM Sleep Behavior Disorder Screening Questionnaire score > 5 points. Logistic regression and Machine Learning (ML) algorithms were used to relate Single Nucleotide Polymorphism (SNPs) in PD-related genes with pRBD. We included 330 PD patients fulfilling all inclusion and exclusion criteria. The final logistic multivariate model revealed that the following SNPs increased the risk of pRBD: GBA_N370S_rs76763715 (OR, 95% CI: 3.38, 1.45–7.93), SNCA_A53T_rs104893877 (8.21, 2.26–36.34), ANK2. CAMK2D_rs78738012 (2.12, 1.08–4.10), and ZNF184_rs9468199 (1.89, 1.08–3.33). Conversely, SNP COQ7. SYT17_rs11343 reduced pRBD risk (0.36, 0.15–0.78). The ML algorithms led to similar results. The predictive models were highly specific (95–99%) but lacked sensitivity (9–39%). We found a distinctive genetic signature for pRBD in PD. The high specificity and low sensitivity of the predictive models suggest that genetic mutations are necessary but not sufficient to develop pRBD in PD. Additional investigations are needed.

1. Introduction

Parkinson’s is the second most frequent neurodegenerative disorder worldwide [1]. Patients are affected by motor and non-motor symptoms, with the latter being the most disturbing [2]. Its main motor symptoms are bradykinesia, resting tremor, rigidity, and postural abnormalities [3]. Patients are also affected by non-motor symptoms, including mood disorders, trouble sleeping, dysautonomia, cognitive dysfunction, and pain, among others [3]. Sleep disorders affect PD patients frequently and negatively impact on quality of life [4,5]. Insufficient sleep interferes with routine activities and can also aggravate motor symptoms in PD. Some of the most frequent disorders are REM sleep behavior disorder (RBD), insomnia, restless legs syndrome (RLS) and periodic limb movement disorder, circadian rhythm sleep disorders, nocturia, sleep-disordered breathing (SBD), and excessive daytime sleepiness.
Compared with the general population, RBD is more prevalent in PD patients, affecting between 20% and 75% of patients [6,7,8,9]. Furthermore, RBD often precedes the onset of motor symptoms in PD by several years and can serve as an early marker or prodromal disease symptom [10]. However, RBD may also develop after PD onset [11].
PD can be sporadic or familial when autosomal mutations are present [12,13]. However, recent evidence indicates that genetic mutations also contribute in non-negligible ways to sporadic PD [14]. A recent meta-analysis of genome-wide association studies (GWAS) included the analysis of 7.8 M SNPs in 37.7 K cases, 18.6 K UK Biobank proxy-cases (having a first-degree relative with PD), and 1.4 M controls [14]. The authors could identify 90 variants that explained 16–36% of the heritable risk of PD depending on prevalence. Interestingly, the presence of some mutations determines distinct phenotypes. For example, in PD patients with mutations in the Leucine-rich repeat kinase 2 (LRRK2) gene, the disease progresses slower than those without mutations and are less frequently affected by non-motor symptoms, including olfaction, REM-sleep behavior disorders, and cognitive dysfunction [15]. PD may also relate to mutations in the GBA gene, encoding the lysosomal enzyme glucocerebrosidase (Gcase) [14]. A recent meta-analysis has shown that PD patients with GBA mutations suffer from accelerated progression of the disease, with more frequent motor fluctuations, depression, and dementia compared to non-affected patients [16].
RBD is a sleep disorder characterized by the loss of muscle atonia during rapid eye movement (REM) sleep, resulting in individuals physically acting out their dreams [17,18]. In individuals with RBD, muscle atonia typical of REM sleep is incomplete or gone, leading to dream-enactment behaviors. These range from mild—simple limb twitches or gestures—to more vigorous and potentially harmful actions like kicking, punching, or even jumping out of bed [17,18]. RBD has been linked to neurodegenerative disorders and synucleinopathies, in particular, Parkinson’s disease (PD), multiple system atrophy, and dementia with Lewy bodies [19].
A disruption in the normal inhibition of muscle activity during REM sleep causes RBD [20,21,22,23]. Individuals with RBD fail at maintaining the typical muscle atonia during REM sleep and then enact dream-related behaviors. The pontomedullary reticular formation, including the subcoeruleus nucleus and other brainstem regions, plays a crucial role in generating REM sleep atonia [23]. Dysfunction or degeneration of these regions disrupts the inhibitory signals sent to the spinal motor neurons, disinhibiting motor activity during REM sleep.
The genetics of idiopathic RBD have been widely studied. In a large genome-wide association study in RBD patients, six RBD-associated loci in five genomic regions were identified: two loci near SCARB2 and INPP5F and three previously reported loci near SNCA, GBA, and TMEM175 [24]. In another case-control study involving 1072 idiopathic RBD patients and 9505 controls, Single Nucleotide Polymorphisms (SNPs) in the HLA-DRB1 were the only genetic factors predicting RBD [25]. SNPs in BST1 and LAMP3 have also been connected with RBD in a study involving 1039 patients and 1852 controls [26]. GBA variants also robustly and differentially increase the risk of idiopathic RBD in a study involving 4147 individuals [27]. The involvement of GBA variants was also associated with RBD in other studies [28,29]. Carriers of the LRRK2 p.N551K-p.R1398H-p.K1423K haplotype have a reduced risk of developing RBD [30]. Conversely, SNPs in the NPC1, which encodes a lysosomal protein involved in cholesterol transport [31], and in the sphingomyelin phosphodiesterase 1 (SMPD1) gene did not show any association with idiopathic RBD [32]. PD-related variants in LRRK2 were also not related to RBD [33,34,35].
Patients who develop PD usually have a more malignant phenotype with a faster rate of disease progression [11]. Indeed, PD patients with RBD (PD-RBD) display higher UPDRS-III motor scores and Hoehn–Yahr scores [9,36], depression, anxiety, apathy, fatigue [11], sleepiness, and cognitive impairment [28].
The reasons for the worst outcome in PD-RBD are currently unknown. We hypothesized that a differential genetic signature might contribute to the torpid evolution of the disease. Therefore, we set out to compare the rate of genetic mutations in patients with PD and probable RBD vs. patients without RBD.

2. Materials and Methods

2.1. Participants

The Parkinson’s Progression Markers Initiative (PPMI) is an ongoing multicenter observational study focused on identifying disease biomarkers in PD patients attending clinical centers from all over the world [37]. The overall goal of PPMI is to identify markers of disease progression to accelerate therapeutic trials to reduce progression of PD disability. All participants signed written informed consent, and the review board of each center approved the protocol. Information is de-identified and shared with involved and uninvolved investigators. We extracted information only from each participant’s baseline visit.
For our study, we selected patients with a clinical diagnosis of idiopathic PD based on the UKPDBBS or the MDS criteria. Patients with missing data, demented patients, those whose PD diagnosis was changed during the first five years after the diagnosis of PD, or those with psychiatric conditions, were excluded.

2.2. Assessment of Probable REM Sleep Behavior Disorder

The “REM Sleep Behavior Disorder Screening Questionnaire” (RBDSQ) was used to screen RBD symptoms [38]. The RBDSQ is a 10-item patient self-rating questionnaire (maximum total score of 13 points) covering the clinical features of RBD, including dream content, nocturnal movements, injuries to self or bed partner, types of motor behaviors during the night, nocturnal awakenings, sleep disruption, and the presence of neurological diseases. The scale was originally developed in English and German, but it has been translated into other languages [39]. Administration time is typically 5 min.
Sensitivity and specificity for polysomnographic-confirmed RBD are 0.96 and 0.56, respectively, considering an RBDSQ score of five points as a positive result [38]. Therefore, we classified PD patients as probable RBD (pRBD) if the RBDSQ score was above 5 points.

2.3. Genomic Data Processing

As part of the screening or baseline visit, blood was drawn, and whole-genome sequencing was performed using a Macrogen Inc. (Seoul, South Korea) sequencer on whole blood-extracted DNA samples [37]. One microgram of each DNA sample was fragmented with Covaris System and prepared following the Illumina TruSeq DNA Sample preparation guide to obtain a final library of 300–400 bp average insert size. Libraries were multiplexed and sequenced on the Illumina HiSeq X platform. Paired-end read sequences were aligned to the GRCh37-hs37d5 genome using the Burrows–Wheeler aligner-maximal exact matches algorithm (BWA-MEM v0.7.13). The Bamsormadup2 tool (v2.0.87) was used to filter duplicates and sort aligned bam files. After filtering duplicated read sequences, the reads were realigned and recalibrated using the GATK pipeline (v3.5). Haplotype caller in the GATK pipeline was used to call variants, including single nucleotide variants (SNVs) and small In/Dels, and to generate genome VCFs. Using the hg38 aligned cohort VCF files from the whole-genome sequencing data, genotype information was extracted using BCF tools and PLINK. We considered the alleles of the 72 variants available in the PPMI database that are associated with an increased PD risk, as identified in a recent large case-control study [14]. We focused on SNPs with a minimum call rate of 95%, a minor allele frequency (MAF) > 1%, and Hardy–Weinberg equilibrium p-values > 0.05.

2.4. Statistical Analysis

Numerical variables were expressed as means ± standard deviation and the categorical ones in percentages. Differences between PD patients with and without NOH were analyzed with a T-test or Chi-square test. We used a logistic regression model to identify SNPs independently associated with probable RBD. We used the Akaike Information Component (AIC) to determine the genetic model that best fitted the data. In the “dominant” model, having one copy of the SNP modified the risk of the outcome. According to the “recessive” model, the risk of the outcome was only modified by the presence of the SNP in both alleles. In the “additive” model, having one or two copies of the SNP affected differently the risk of the outcome. For the multivariate model, we selected the one with the highest AIC. The Benjamini and Hochberg step-up procedure was used to control the False Discovery Rate and adjust the p-values. All analyses were performed using R Statistical Software (v4.1.2; R Core Team 2021).

2.5. Machine-Learning Models

We used Machine-Learning (ML) algorithms to further model the relationship between probable RBD with SNPs and other covariates. We fitted Logistic regression, Bayes Naïve, Decision trees, Boosted Decision trees, Neural Networks, Support-Vector Machines (SVM), and Random Forest models to a “development” and “validation” subsamples comprising 70%/30% of the original sample. For each model, we computed the Area Under the Curve of the Receiver–Operator Curve (AUC-ROC) and the sensitivity and specificity for detecting pRBD. In the second row of analyses, we included clinical covariates in the models. We also used Leave-one-out CV (LOOCV) as the resampling technique, aiming at maximizing the size of the development sub-sample. In this cross-validation technique, ML models are fitted to the whole sample minus 1 participant, who serves to test the model validity. This procedure is iterated through the whole sample. Therefore, both the development and validation subsamples included all participants. All analyses were performed with R, using the following libraries: rstatix, e1071, C50, NeuralNet, Kernlab, RandomForest, and caret.

3. Results

3.1. Characteristics of the Sample

Six hundred and seventy-four patients fulfilled the inclusion criteria. The final sample included 330 PD patients, as 344 had missing data. Characteristics of patients with pRBD or no RBD are shown in Table 1. Most notably, patients with pRBD had a younger disease onset, a longer disease duration, more severe motor symptoms, more frequent antiparkinsonian treatment, and more frequent motor fluctuations.

3.2. Genetic Factors Connected with RBD

We observed significant differences between PD patients with pRBD and without RBD in 11 SNPs (Table 2). The final multivariate model revealed that the SNP GBA_N370S_rs76763715, SNCA_A53T_rs104893877, ANK2.CAMK2D_rs78738012, ZNF184_rs9468199 increased the risk of pRBD. Conversely, the SNP COQ7.SYT17_rs11343 reduced pRBD risk.

3.3. Machine-Learning Models

We fitted a series of ML models to corroborate these findings. Table 3 shows the performance of ML models in the “validation” subsample, which comprised 1/3 of the original sample. The logistic regression and Bayes Naïve algorithms showed a sensitivity of 39% and a specificity of 95%. The Neural Network model had the highest AUC-ROC; however, sensitivity and specificity were not different from the logistic regression or Bayes Naïve models.
To increase the predictive power of the ML models, we included Levodopa-Equivalent Daily Dose, MDS-UPDRS I + II + III, and Motor Fluctuations, which show differences in patients with pRBD or no RBD (Table 1). Inclusion of Age at Disease Onset and Disease Duration was not possible due to missing data (Table 1). The models’ performance and coefficients for the variables included are shown in Table 4. The most significant change was a 10% increase in sensitivity, which was lost when the LOOCV method was used to build the ML model.

4. Discussion

We hypothesized that PD patients with pRBD may have a different genetic background as compared with PD patients without RBD. In this study, we observed that pRBD was directly associated with GBA_N370S_rs76763715, SNCA_A53T_rs104893877, and ANK2.CAMK2D_rs78738012, ZNF184_rs9468199, and indirectly, with SNP COQ7.SYT17_rs11343. These results support our initial hypothesis, shedding light on the possible origin of the worst prognosis observed in patients with RBD.
Before further discussing these findings, the lack of polysomnographic confirmation of RBD diagnosis deserves a few words. Both the absence of muscle atonia during REM sleep and the presence of dream-enactment behaviors during polysomnography are required to confirm RBD diagnosis [40]. The differential diagnosis of RBD includes non-REM parasomnia (confusional arousals, sleepwalking, sleep terrors), nightmares, benign sleeptalking, nocturnal frontal lobe epilepsy, and sleep-fragmenting conditions like obstructive sleep apnea and periodic limb movements, among others [38]. As polysomnography is impractical for large multicentric studies, surveys are commonly used [9]. The RBDSQ used here is highly sensitive but unspecific. A negative result drastically reduced RBD likelihood, while a positive result should be confirmed by PSG, which was not done in this case. However, there is no theoretical background to think that RBD differential diagnoses are related to the SNPs assessed in this study. Therefore, the relationships between RBD and the SNPs observed were probably underestimated due to the presence of non-RBD patients among those with pRBD, and less strong relationships may have been missed out. Further research is needed studying confirmed RBD patients. Finally, we didn’t assess the “codominant” model—when the two alleles are expressed separately, yielding different effects. The database did not include the data needed to assess this model.
Several studies found that GBA and SNCA SNPs are more frequent among patients with idiopathic RBD compared to controls. In line with these findings, we observed that GBA_N370S_rs76763715 and SNCA_A53T_rs104893877 related to more frequent pRBD among people living with PD. Compared with non-carriers, PD patients with SNCA variants have more frequent non-motor symptoms, including RBD, and a similar disease progression rate (SNPs rs2870004, rs356182, rs5019538, and rs763443) [41]. Some GBA variants, like rs2230288/E326K, rs75548401/T369M, and rs369068553/V460L, are also linked to more frequent non-motor symptoms, including RBD, but also to a more aggressive motor disease [42,43,44]. Therefore, the presence of genetic mutations in the SNCA gene may explain the worse prognosis in PD-RBD.
A study of Chinese PD patients showed that the variant rs9468199 in the ZNF184 gene was associated with more frequent RBD [45]. The ZNF184 encodes a Kruppel C2H2-type zinc-finger protein family member, which participates in gene expression regulation. One study found that REM sleep deprivation in rats induced significant ZNF184 down-regulation in the brain [46]. The effects of this mutation on other features of PD are unknown.
We report for the first time the associations between pRBD and ANK2.CAMK2D_rs78738012 and COQ7.SYT17_rs11343 variants. The ANK2 gene encodes a member of the ankyrin family of proteins, linking the integral membrane proteins to the underlying spectrin–actin cytoskeleton. Ankyrins play key roles in organizing the axon initial segment and nodes of Ranvier, and they organize and stabilize neurotransmitter receptors, modulate dendritic spine morphology, and control adhesion to the presynaptic site [47]. The product of the CAMK2 belongs to the serine/threonine protein kinase family and to the Ca (2+)/calmodulin-dependent protein kinase subfamily. Calcium signaling is crucial for several aspects of plasticity at glutamatergic synapses. Interestingly, Camk2b knockout mice show reduced sleep [48]. The phosphorylation states of CaMKIIβ appear to control sleep induction and maintenance processes differently.
The SYT17 encodes the Synaptotagmin 17 protein, which has calcium ion binding activity, phospholipid binding activity, and syntaxin binding activity. It is involved in the positive regulation of dendrite extension. The suprachiasmatic nucleus—the master biological clock of the brain—expresses high levels of SYT17 [49]. Several findings suggest that SYT17 is involved in regulating circadian rhythms. The best-fitting model for the risk of this gene was the “recessive” model, suggesting that the SNPs might be linked to a loss of gene function. Therefore, only when both alleles are compromised protein expression might fall below critical levels. Further research is needed to assess these factors’ importance in RBD pathophysiology and their impact on other features of PD.
Our predictive models were highly specific but poorly sensitive. Namely, PD patients without pathogenic SNPs showed a minimal risk of suffering from RBD. PD patients with pathogenic mutations had a moderate increase in the risk of pRBD. These findings suggest that genetic background is necessary for the development of pRBD but not sufficient. The non-genetic factors that might contribute to the development of idiopathic RBD include smoking, previous head injury, fewer years of formal schooling, and working on a farm [50]. However, the risk factors for pRBD have been less frequently studied. The inclusion of clinical variables like PD severity, levodopa-equivalent daily dose, or the presence of motor fluctuation in the logistic predictive model increased sensitivity by 10%. These factors might contribute to, while others still unknown, are necessary to develop pRBD.
Logistic regression is the cornerstone of multivariate testing and is widely used in epidemiological studies [51,52,53]. The recent development in ML algorithms may provide more sophisticated and powerful methods for detecting associations between variables in epidemiological studies. We compared the results of several ML algorithms with regression logistics in our dataset. More sophisticated algorithms did not outperform logistic regression, emphasizing the power of this old technique.

5. Conclusions

We found a distinctive genetic signature for pRBD in PD. SNPs in genes GBA, SNCA, ANK2.CAMK2D and ZNF184 increased while in COQ7.SYT17 reduced the risk of pRBD. Genetic mutations appear necessary but not sufficient to develop RBD. Unaccounted factors remain obscure and additional studies are needed.

6. Future Directions

We hypothesized that the more torpid evolution of patients with PD and RBD could result from a genetic mutation. We observed that PD patients with pRBD had an increased frequency of mutations in the GBA and SNCA genes, which relate to a worse prognosis. We also observed mutations in the ANK2.CAMK2D and ZNF184 genes increased the risk of pRBD in PD, while mutations in the COQ7.SYT17 reduced it. Further research is needed to evaluate the pathophysiological implications of these mutations. Furthermore, they might also represent therapeutic targets worth studying.
The main limitation of this study, which should be addressed in future studies, is the inclusion of polysomnography-unconfirmed cases of RBD. The lack of polysomnographic confirmation and the relatively low sample size might have limited our ability to find mutations significantly associated with RBD. In other words, we might have missed some mutations weakly related to RBD. Finally, future studies should address the presence and effects of co-dominant mutations.

Author Contributions

Conceptualization, G.C. and S.P.-L.; methodology, G.C., S.B., L.U. and H.B.; statistical analysis, G.C. and S.P.-L.; writing—review and editing, M.O.-L., G.C., S.B., H.B., L.U. and S.P.-L.; supervision, S.P.-L. and F.C. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data used in the preparation of this article were obtained from the Parkinson’s Progression Markers Initiative (PPMI) database (www.ppmi-info.org/data, accessed on 3 April 2023). For up-to-date information on the study, visit www.ppmi-info.org (accessed on 3 April 2023).

Acknowledgments

PPMI—a public-private partnership—is funded by the Michael J. Fox Foundation for Parkinson’s Research funding partners 4D Pharma, Abbvie, Acurex Therapeutics, Allergan, Amathus Therapeutics, ASAP, Avid Radiopharmaceuticals, Bial Biotech, Biogen, BioLegend, Bristol-Myers Squibb, Calico, Celgene, Dacapo Brain Science, Denali, The Edmond J. Safra Foundation, GE Healthcare, Genentech, GlaxoSmithKline, Golub Capital, Handl Therapeutics, Insitro, Janssen Neuroscience, Lilly, Lundbeck, Merck, Meso Scale Discovery, Neurocrine Biosciences, Pfizer, Piramal, Prevail, Roche, Sanofi Genzyme, Servier, Takeda, Teva, UCB, Verily, and Voyager Therapeutics. The authors did not receive any other funding for this study.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Dorsey, E.R.; Constantinescu, R.; Thompson, J.P.; Biglan, K.M.; Holloway, R.G.; Kieburtz, K.; Marshall, F.J.; Ravina, B.M.; Schifitto, G.; Siderowf, A.; et al. Projected number of people with Parkinson disease in the most populous nations, 2005 through 2030. Neurology 2007, 68, 384–386. [Google Scholar] [CrossRef] [PubMed]
  2. Lang, A.E.; Espay, A.J. Disease Modification in Parkinson’s Disease: Current Approaches, Challenges, and Future Considerations. Mov. Disord. 2018, 33, 660–677. [Google Scholar] [CrossRef]
  3. Chaudhuri, K.R.; Schapira, A.H. Non-motor symptoms of Parkinson’s disease: Dopaminergic pathophysiology and treatment. Lancet Neurol. 2009, 8, 464–474. [Google Scholar] [CrossRef] [PubMed]
  4. Avidan, A.; Hays, R.D.; Diaz, N.; Bordelon, Y.; Thompson, A.W.; Vassar, S.D.; Vickrey, B.G. Associations of sleep disturbance symptoms with health-related quality of life in Parkinson’s disease. J. Neuropsychiatry Clin. Neurosci. 2013, 25, 319–326. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Gallagher, D.A.; Lees, A.J.; Schrag, A. What are the most important nonmotor symptoms in patients with Parkinson’s disease and are we missing them? Mov. Disord. 2010, 25, 2493–2500. [Google Scholar] [CrossRef]
  6. Zhang, J.; Xu, C.Y.; Liu, J. Meta-analysis on the prevalence of REM sleep behavior disorder symptoms in Parkinson’s disease. BMC Neurol. 2017, 17, 23. [Google Scholar] [CrossRef] [Green Version]
  7. Baumann-Vogel, H.; Hor, H.; Poryazova, R.; Valko, P.; Werth, E.; Baumann, C.R. REM sleep behavior in Parkinson disease: Frequent, particularly with higher age. PLoS ONE 2020, 15, e0243454. [Google Scholar] [CrossRef]
  8. Hu, M.T. REM sleep behavior disorder (RBD). Neurobiol. Dis. 2020, 143, 104996. [Google Scholar] [CrossRef]
  9. Barasa, A.; Wang, J.; Dewey, R.B., Jr. Probable REM Sleep Behavior Disorder Is a Risk Factor for Symptom Progression in Parkinson Disease. Front. Neurol. 2021, 12, 651157. [Google Scholar] [CrossRef]
  10. Ferri, R.; Cosentino, F.I.; Pizza, F.; Aricò, D.; Plazzi, G. The timing between REM sleep behavior disorder and Parkinson’s disease. Sleep Breath. 2014, 18, 319–323. [Google Scholar] [CrossRef]
  11. Neikrug, A.B.; Avanzino, J.A.; Liu, L.; Maglione, J.E.; Natarajan, L.; Corey-Bloom, J.; Palmer, B.W.; Loredo, J.S.; Ancoli-Israel, S. Parkinson’s disease and REM sleep behavior disorder result in increased non-motor symptoms. Sleep Med. 2014, 15, 959–966. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Singleton, A.B.; Farrer, M.; Johnson, J.; Singleton, A.; Hague, S.; Kachergus, J.; Hulihan, M.; Peuralinna, T.; Dutra, A.; Nussbaum, R.; et al. alpha-Synuclein locus triplication causes Parkinson’s disease. Science 2003, 302, 841. [Google Scholar] [CrossRef] [Green Version]
  13. Polymeropoulos, M.H.; Higgins, J.J.; Golbe, L.I.; Johnson, W.G.; Ide, S.E.; Di Iorio, G.; Sanges, G.; Stenroos, E.S.; Pho, L.T.; Schaffer, A.A.; et al. Mapping of a gene for Parkinson’s disease to chromosome 4q21-q23. Science 1996, 274, 1197–1199. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Nalls, M.A.; Blauwendraat, C.; Vallerga, C.L.; Heilbron, K.; Bandres-Ciga, S.; Chang, D.; Tan, M.; Kia, D.A.; Noyce, A.J.; Xue, A.; et al. Identification of novel risk loci, causal insights, and heritable risk for Parkinson’s disease: A meta-analysis of genome-wide association studies. Lancet Neurol. 2019, 18, 1091–1102. [Google Scholar] [CrossRef] [PubMed]
  15. Tolosa, E.; Vila, M.; Klein, C.; Rascol, O. LRRK2 in Parkinson disease: Challenges of clinical trials. Nat. Rev. Neurol. 2020, 16, 97–107. [Google Scholar] [CrossRef]
  16. Zhang, Y.; Shu, L.; Zhou, X.; Pan, H.; Xu, Q.; Guo, J.; Tang, B.; Sun, Q. A Meta-Analysis of GBA-Related Clinical Symptoms in Parkinson’s Disease. Parkinsons Dis. 2018, 2018, 3136415. [Google Scholar] [CrossRef] [Green Version]
  17. Boeve, B.F. REM sleep behavior disorder: Updated review of the core features, the REM sleep behavior disorder-neurodegenerative disease association, evolving concepts, controversies, and future directions. Ann. N. Y. Acad. Sci. 2010, 1184, 15–54. [Google Scholar] [CrossRef] [Green Version]
  18. St Louis, E.K.; Boeve, B.F. REM Sleep Behavior Disorder: Diagnosis, Clinical Implications, and Future Directions. Mayo Clin. Proc. 2017, 92, 1723–1736. [Google Scholar] [CrossRef]
  19. Boeve, B.F.; Silber, M.H.; Ferman, T.J.; Lucas, J.A.; Parisi, J.E. Association of REM sleep behavior disorder and neurodegenerative disease may reflect an underlying synucleinopathy. Mov. Disord. 2001, 16, 622–630. [Google Scholar] [CrossRef]
  20. Boeve, B.F. Idiopathic REM sleep behaviour disorder in the development of Parkinson’s disease. Lancet Neurol. 2013, 12, 469–482. [Google Scholar] [CrossRef] [Green Version]
  21. Iranzo, A.; Santamaria, J.; Tolosa, E. The clinical and pathophysiological relevance of REM sleep behavior disorder in neurodegenerative diseases. Sleep Med. Rev. 2009, 13, 385–401. [Google Scholar] [CrossRef]
  22. McCarter, S.J.; St Louis, E.K.; Boeve, B.F. REM sleep behavior disorder and REM sleep without atonia as an early manifestation of degenerative neurological disease. Curr. Neurol. Neurosci. Rep. 2012, 12, 182–192. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. St Louis, E.K.; McCarter, S.J.; Boeve, B.F.; Silber, M.H.; Kantarci, K.; Benarroch, E.E.; Rando, A.; Tippmann-Peikert, M.; Olson, E.J.; Mauermann, M.L. Lesional REM sleep behavior disorder localizes to the dorsomedial pons. Neurology 2014, 83, 1871–1873. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Krohn, L.; Heilbron, K.; Blauwendraat, C.; Reynolds, R.H.; Yu, E.; Senkevich, K.; Rudakou, U.; Estiar, M.A.; Gustavsson, E.K.; Brolin, K.; et al. Genome-wide association study of REM sleep behavior disorder identifies polygenic risk and brain expression effects. Nat. Commun. 2022, 13, 7496. [Google Scholar] [CrossRef]
  25. Yu, E.; Krohn, L.; Ruskey, J.A.; Asayesh, F.; Spiegelman, D.; Shah, Z.; Chia, R.; Arnulf, I.; Hu, M.T.M.; Montplaisir, J.Y.; et al. HLA in isolated REM sleep behavior disorder and Lewy body dementia. medRxiv 2023. [Google Scholar] [CrossRef]
  26. Mufti, K.; Yu, E.; Rudakou, U.; Krohn, L.; Ruskey, J.A.; Asayesh, F.; Laurent, S.B.; Spiegelman, D.; Arnulf, I.; Hu, M.T.M.; et al. Novel Associations of BST1 and LAMP3 With REM Sleep Behavior Disorder. Neurology 2021, 96, e1402–e1412. [Google Scholar] [CrossRef] [PubMed]
  27. Krohn, L.; Ruskey, J.A.; Rudakou, U.; Leveille, E.; Asayesh, F.; Hu, M.T.M.; Arnulf, I.; Dauvilliers, Y.; Högl, B.; Stefani, A.; et al. GBA variants in REM sleep behavior disorder: A multicenter study. Neurology 2020, 95, e1008–e1016. [Google Scholar] [CrossRef]
  28. Rolinski, M.; Szewczyk-Krolikowski, K.; Tomlinson, P.R.; Nithi, K.; Talbot, K.; Ben-Shlomo, Y.; Hu, M.T. REM sleep behaviour disorder is associated with worse quality of life and other non-motor features in early Parkinson’s disease. J. Neurol. Neurosurg. Psychiatry 2014, 85, 560–566. [Google Scholar] [CrossRef] [Green Version]
  29. Thaler, A.; Bregman, N.; Gurevich, T.; Shiner, T.; Dror, Y.; Zmira, O.; Gan-Or, Z.; Bar-Shira, A.; Gana-Weisz, M.; Orr-Urtreger, A.; et al. Parkinson’s disease phenotype is influenced by the severity of the mutations in the GBA gene. Parkinsonism Relat. Disord. 2018, 55, 45–49. [Google Scholar] [CrossRef]
  30. Ouled Amar Bencheikh, B.; Ruskey, J.A.; Arnulf, I.; Dauvilliers, Y.; Monaca, C.C.; De Cock, V.C.; Gagnon, J.F.; Spiegelman, D.; Hu, M.T.M.; Högl, B.; et al. LRRK2 protective haplotype and full sequencing study in REM sleep behavior disorder. Parkinsonism Relat. Disord. 2018, 52, 98–101. [Google Scholar] [CrossRef]
  31. Somerville, E.N.; Krohn, L.; Yu, E.; Rudakou, U.; Senkevich, K.; Ruskey, J.A.; Asayesh, F.; Ahmad, J.; Spiegelman, D.; Dauvilliers, Y.; et al. NPC1 variants are not associated with Parkinson’s disease, REM-sleep behavior disorder or dementia with Lewy bodies in European cohorts. Neurobiol. Aging 2023, 127, 94–98. [Google Scholar] [CrossRef] [PubMed]
  32. Rudakou, U.; Futhey, N.C.; Krohn, L.; Ruskey, J.A.; Heilbron, K.; Cannon, P.; Alam, A.; Arnulf, I.; Hu, M.T.M.; Montplaisir, J.Y.; et al. SMPD1 variants do not have a major role in rapid eye movement sleep behavior disorder. Neurobiol. Aging 2020, 93, 142.e5–142.e7. [Google Scholar] [CrossRef] [PubMed]
  33. Barber, T.R.; Lawton, M.; Rolinski, M.; Evetts, S.; Baig, F.; Ruffmann, C.; Gornall, A.; Klein, J.C.; Lo, C.; Dennis, G.; et al. Prodromal Parkinsonism and Neurodegenerative Risk Stratification in REM Sleep Behavior Disorder. Sleep 2017, 40, zsx071. [Google Scholar] [CrossRef] [Green Version]
  34. Fernández-Santiago, R.; Iranzo, A.; Gaig, C.; Serradell, M.; Fernández, M.; Tolosa, E.; Santamaría, J.; Ezquerra, M. Absence of LRRK2 mutations in a cohort of patients with idiopathic REM sleep behavior disorder. Neurology 2016, 86, 1072–1073. [Google Scholar] [CrossRef] [PubMed]
  35. Kestenbaum, M.; Alcalay, R.N. Clinical Features of LRRK2 Carriers with Parkinson’s Disease. Adv. Neurobiol. 2017, 14, 31–48. [Google Scholar] [CrossRef]
  36. Zhu, R.L.; Xie, C.J.; Hu, P.P.; Wang, K. Clinical variations in Parkinson’s disease patients with or without REM sleep behaviour disorder: A meta-analysis. Sci. Rep. 2017, 7, 40779. [Google Scholar] [CrossRef] [Green Version]
  37. Parkinson Progression Marker Initiative. The Parkinson Progression Marker Initiative (PPMI). Prog. Neurobiol. 2011, 95, 629–635. [Google Scholar] [CrossRef]
  38. Stiasny-Kolster, K.; Mayer, G.; Schäfer, S.; Möller, J.C.; Heinzel-Gutenbrunner, M.; Oertel, W.H. The REM sleep behavior disorder screening questionnaire--a new diagnostic instrument. Mov. Disord. 2007, 22, 2386–2393. [Google Scholar] [CrossRef]
  39. Skorvanek, M.; Feketeova, E.; Kurtis, M.M.; Rusz, J.; Sonka, K. Accuracy of Rating Scales and Clinical Measures for Screening of Rapid Eye Movement Sleep Behavior Disorder and for Predicting Conversion to Parkinson’s Disease and Other Synucleinopathies. Front. Neurol. 2018, 9, 376. [Google Scholar] [CrossRef]
  40. Högl, B.; Stefani, A. REM sleep behavior disorder (RBD): Update on diagnosis and treatment. Somnologie 2017, 21, 1–8. [Google Scholar] [CrossRef] [Green Version]
  41. Szwedo, A.A.; Pedersen, C.C.; Ushakova, A.; Forsgren, L.; Tysnes, O.B.; Counsell, C.E.; Alves, G.; Lange, J.; Macleod, A.D.; Maple-Grødem, J. Association of SNCA Parkinson’s Disease Risk Polymorphisms With Disease Progression in Newly Diagnosed Patients. Front. Neurol. 2020, 11, 620585. [Google Scholar] [CrossRef]
  42. Höglinger, G.; Schulte, C.; Jost, W.H.; Storch, A.; Woitalla, D.; Krüger, R.; Falkenburger, B.; Brockmann, K. GBA-associated PD: Chances and obstacles for targeted treatment strategies. J. Neural Transm. 2022, 129, 1219–1233. [Google Scholar] [CrossRef] [PubMed]
  43. Maple-Grødem, J.; Dalen, I.; Tysnes, O.B.; Macleod, A.D.; Forsgren, L.; Counsell, C.E.; Alves, G. Association of GBA Genotype With Motor and Functional Decline in Patients With Newly Diagnosed Parkinson Disease. Neurology 2021, 96, e1036–e1044. [Google Scholar] [CrossRef] [PubMed]
  44. Yahalom, G.; Greenbaum, L.; Israeli-Korn, S.; Fay-Karmon, T.; Livneh, V.; Ruskey, J.A.; Roncière, L.; Alam, A.; Gan-Or, Z.; Hassin-Baer, S. Carriers of both GBA and LRRK2 mutations, compared to carriers of either, in Parkinson’s disease: Risk estimates and genotype-phenotype correlations. Parkinsonism Relat. Disord. 2019, 62, 179–184. [Google Scholar] [CrossRef]
  45. Zhang, B.; Cui, C.; Yu, H.; Li, G. Association between ZNF184 and symptoms of Parkinson’s disease in southern Chinese. Neurol. Sci. 2020, 41, 2121–2126. [Google Scholar] [CrossRef] [PubMed]
  46. Pandey, A.; Oliver, R.; Kar, S.K. Differential Gene Expression in Brain and Liver Tissue of Wistar Rats after Rapid Eye Movement Sleep Deprivation. Clocks Sleep 2020, 2, 442–465. [Google Scholar] [CrossRef] [PubMed]
  47. Smith, K.R.; Penzes, P. Ankyrins: Roles in synaptic biology and pathology. Mol. Cell. Neurosci. 2018, 91, 131–139. [Google Scholar] [CrossRef]
  48. Tone, D.; Ode, K.L.; Zhang, Q.; Fujishima, H.; Yamada, R.G.; Nagashima, Y.; Matsumoto, K.; Wen, Z.; Yoshida, S.Y.; Mitani, T.T.; et al. Distinct phosphorylation states of mammalian CaMKIIβ control the induction and maintenance of sleep. PLoS Biol. 2022, 20, e3001813. [Google Scholar] [CrossRef]
  49. Fujioka, A.; Nagano, M.; Ikegami, K.; Masumoto, K.H.; Yoshikawa, T.; Koinuma, S.; Nakahama, K.I.; Shigeyoshi, Y. Circadian expression and specific localization of synaptotagmin17 in the suprachiasmatic nucleus, the master circadian oscillator in mammals. Brain Res. 2023, 1798, 148129. [Google Scholar] [CrossRef]
  50. Postuma, R.B.; Montplaisir, J.Y.; Pelletier, A.; Dauvilliers, Y.; Oertel, W.; Iranzo, A.; Ferini-Strambi, L.; Arnulf, I.; Hogl, B.; Manni, R.; et al. Environmental risk factors for REM sleep behavior disorder: A multicenter case-control study. Neurology 2012, 79, 428–434. [Google Scholar] [CrossRef] [Green Version]
  51. Bagley, S.C.; White, H.; Golomb, B.A. Logistic regression in the medical literature: Standards for use and reporting, with particular attention to one medical domain. J. Clin. Epidemiol. 2001, 54, 979–985. [Google Scholar] [CrossRef] [PubMed]
  52. Kleinbaum, D.G.; Kupper, L.L.; Chambless, L.E. Logistic regression analysis of epidemiologic data: Theory and practice. Commun. Stat.—Theory Methods 1982, 11, 485–547. [Google Scholar] [CrossRef]
  53. Sperandei, S. Understanding logistic regression analysis. Biochem. Med. 2014, 24, 12–18. [Google Scholar] [CrossRef] [PubMed]
Table 1. Characteristics of PD patients with probable REM-sleep behavior disorder (pRBD) or no RBD.
Table 1. Characteristics of PD patients with probable REM-sleep behavior disorder (pRBD) or no RBD.
CharacteristicOverall
(N = 330)
No RBD
(N = 240)
pRBD
(N = 90)
p-Value
Years of education 0.008
Mean (SD)15.6 (3.77)15.9 (3.59)14.6 (4.12)
Median (Min, Max)16.0 (0, 26.0)16.0 (0, 25.0)15.0 (4.00, 26.0)
Missing17 (5.1%)11 (4.6%)6 (6.7%)
Sex 0.533
M143 (43.1%)107 (44.6%)36 (40.0%)
F189 (56.9%)133 (55.4%)54 (60.0%)
Age 0.261
Mean (SD)61.2 (10.9)61.6 (10.6)60.1 (11.8)
Median (Min, Max)63.0 (31.0, 84.0)64.0 (31.0, 83.0)63.0 (31.0, 84.0)
Age at PD onset 0.050
Mean (SD)57.8 (10.9)59.3 (11.0)55.2 (10.6)
Median (Min, Max)60.0 (29.0, 80.0)61.0 (32.0, 80.0)58.5 (29.0, 70.0)
Missing211 (63.6%)165 (68.8%)46 (51.1%)
PD duration (months) 0.028
Mean (SD)37.3 (23.9)33.4 (23.6)43.2 (23.1)
Median (Min, Max)31.0 (2.92, 85.0)29.0 (2.92, 80.1)43.1 (4.01, 85.0)
Missing211 (63.6%)165 (68.8%)46 (51.1%)
Antiparkinsonians <0.001
No198 (59.6%)158 (65.8%)40 (44.4%)
Yes134 (40.4%)82 (34.2%)50 (55.6%)
Levodopa <0.001
No241 (72.6%)188 (78.3%)53 (58.9%)
Yes91 (27.4%)52 (21.7%)37 (41.1%)
Dopamine agonists 0.002
No274 (82.5%)209 (87.1%)65 (72.2%)
Yes58 (17.5%)31 (12.9%)25 (27.8%)
MAO-B inhibitors 0.016
No240 (72.3%)183 (76.3%)56 (62.2%)
Yes92 (27.7%)57 (23.8%)34 (37.8%)
Entacapano 0.015
No310 (93.4%)230 (95.8%)79 (87.8%)
Yes22 (6.63%)10 (4.17%)11 (12.2%)
Levodopa equivalent daily dose 0.001
Mean (SD)280 (714)158 (323)564 (1170)
Median (Min, Max)0 (0, 7630)0 (0, 2400)140 (0, 7630)
Elixhauser comorbidities score 0.186
Mean (SD)1.26 (3.14)1.13 (2.85)1.64 (3.83)
Median (Min, Max)0 (0, 20.0)0 (0, 20.0)0 (0, 19.0)
MDS-UPDRS I-III <0.001
Mean (SD)18.4 (22.0)14.4 (17.5)29.5 (28.5)
Median (Min, Max)8.00 (0, 130)7.00 (0, 106)22.0 (0, 130)
Missing8 (2.4%)2 (0.8%)4 (4.4%)
Dyskinesias 0.84
No313 (96.3%)230 (96.6%)83 (95.4%)
Yes12 (3.69%)8 (3.36%)4 (4.60%)
Missing7 (2.1%)2 (0.8%)3 (3.3%)
Motor fluctuations <0.001
No296 (91.1%)226 (95.0%)70 (80.5%)
Yes29 (8.92%)12 (5.04%)17 (19.5%)
Missing7 (2.1%)2 (0.8%)3 (3.3%)
MoCA score 0.360
Mean (SD)27.4 (2.76)27.5 (2.57)27.2 (3.24)
Median (Min, Max)28.0 (11.0, 30.0)28.0 (11.0, 30.0)28.0 (12.0, 30.0)
Missing19 (5.7%)12 (5.0%)7 (7.8%)
Minimal cognitive impairment 0.999
No148 (95.5%)98 (95.1%)50 (96.2%)
Yes7 (4.52%)5 (4.85%)2 (3.85%)
Missing177 (53.3%)137 (57.1%)38 (42.2%)
PD Dementia 0.999
No152 (98.1%)102 (99.0%)50 (96.2%)
Yes3 (1.94%)1 (0.971%)2 (3.85%)
Missing177 (53.3%)137 (57.1%)38 (42.2%)
Table 2. SNPs in patients with probable REM-sleep behavior disorder (pRBD) or no RBD.
Table 2. SNPs in patients with probable REM-sleep behavior disorder (pRBD) or no RBD.
Number of Risk Alleles for Each SNPOverall
(N = 330)
No RBD
(N = 240)
pRBD
(N = 90)
Chi-sq
p-Value
Additive Model
OR (95% CI)
p-Value, (AIC)
Dominant Model
OR (95% CI)
p-Value, (AIC)
Recessive Model
OR (95% CI)
p-Value, (AIC)
Full Model OR (95% CI)
p-Value
FDR p-Value
GBA_N370S_rs76763715 0.004-3.22 (1.48–7.07)-3.38 (1.45–7.93)
0301 (91.3%)226 (94.2%)75 (83.3%) 0.002 (382.14) 0.004
129 (8.73%)14 (5.83%)15 (16.7%) SELECTED 0.018
20 (0%)0 (0%)0 (0%)
GBA_E365K_rs2230288 0.026-7.00 (1.47–49.51)-5.59 (1.05–42.09)
0323 (97.9%)238 (99.2%)85 (94.4%) 0.02 (384.69) 0.050
17 (2.11%)2 (0.833%)5 (5.56%) SELECTED 0.084
20 (0%)0 (0%)0 (0%)
ACMSD.TMEM163_rs6430538 0.0220.64 (0.45–0.89)0.64 (0.39–1.06)0.39 (0.19–0.77)0.48 (0.22–0.99)
0115 (34.9%)77 (32.1%)38 (42.2%) 0.01 (383.93)0.08 (387.81)0.009 (383.03)0.050
1142 (43.1%)101 (42.1%)41 (45.6%) SELECTED0.084
273 (22.0%)62 (25.8%)11 (12.2%)
MCCC1_rs12637471 0.0771.07 (0.69–1.64)0.90 (0.54–1.48)3.35 (0.98–11.92)3.05 (0.83–11.65)
0207 (62.3%)149 (62.1%)58 (64.4%) 0.74 (390.62)0.69 (390.57)0.05 (386.97)0.110
1112 (34.3%)86 (35.8%)26 (28.9%) SELECTED0.110
211 (3.31%)5 (2.08%)6 (6.67%)
FAM47E.STBD1_rs6812193 0.0771.22 (0.84–1.76)1.02 (0.62–1.68)2.14 (1.05–4.27)1.77 (0.79–3.91)
0132 (41.9%)100 (41.7%)37 (41.1%) 0.26 (389.49)0.93 (390.72)0.03 (386.31)0.150
1155 (46.7%)118 (49.2%)37 (41.1%) SELECTED0.170
238 (11.4%)22 (9.17%)16 (17.8%)
SNCA_A53T_rs104893877 <0.001-7.37 (2.39–27.48)-8.21 (2.26–36.34)
0312 (95.8%)236 (98.3%)80 (88.9%) <0.001 (378.33) 0.002
114 (4.22%)4 (1.67%)10 (11.1%) SELECTED 0.014
20 (0%)0 (0%)0 (0%)
ANK2.CAMK2D_rs78738012 0.0052.26 (1.34–3.82)2.47 (1.39–4.36)2.70 (0.32–22.81)2.12 (1.08–4.10)
0265 (80.1%)203 (84.6%)62 (68.9%) 0.002 (381.38)0.001 (381.19)0.32 (389.79)0.030
161 (18.7%)35 (14.6%)26 (28.9%) SELECTED 0.050
24 (1.20%)2 (0.833%)2 (2.22%)
ZNF184_rs9468199 0.0221.58 (1.04–2.41)1.92 (1.17–3.16)0.88 (0.19–3.04)1.89 (1.08–3.33)
0213 (64.8%)165 (68.8%)48 (53.3%) 0.03 (386.15)0.009 (384.07)0.85 (390.70)0.030
1105 (31.6%)66 (27.5%)39 (43.3%) SELECTED 0.050
212 (3.61%)9 (3.75%)3 (3.33%)
CTSB_rs1293298 0.1080.96 (0.64–1.41)0.77 (0.47–1.25)1.87 (0.78–4.30)1.91 (0.73–4.86)
0157 (47.9%)110 (45.8%)47 (52.2%) 0.84 (390.69)0.30 (389.66)0.14 (388.67)0.17
1148 (44.6%)115 (47.9%)33 (36.7%) SELECTED0.17
225 (7.53%)15 (6.25%)10 (11.1%)
COQ7.SYT17_rs11343 0.0070.81 (0.57–1.15)1.19 (0.69–2.10)0.38 (0.17–0.75)0.36 (0.15–0.78)
093 (28.0%)70 (29.2%)23 (25.6%) 0.25 (389.41)0.51 (390.30)0.009 (382.81)0.010
1168 (50.9%)111 (46.3%)57 (63.3%) SELECTED0.041
269 (21.1%)59 (24.6%)10 (11.1%)
ZNF646.KAT8.BCKDK_rs14235 0.071.24 (0.87–1.75)1.73 (1.03–3.01)0.89 (0.45–1.69)1.67 (0.93–3.08)
0117 (35.5%)93 (38.8%)24 (26.7%) 0.22 (389.23)0.04 (386.43)0.74 (390.62)0.09
1158 (47.9%)106 (44.2%)52 (57.8%) SELECTED 0.11
255 (16.6%)41 (17.1%)14 (15.6%)
The Akaike Information Coefficient (AIC) was used to compare the additive, dominant, and recessive models. The one with the lowest AIC was selected for the multivariate model, provided the p-value was <0.15. FDR = False Discovery Rate p-value adjustment by the Benjamini and Hochberg step-up procedure.
Table 3. Performance of ML models for predicting possible RBD in the “validation” subsample.
Table 3. Performance of ML models for predicting possible RBD in the “validation” subsample.
ModelAUC ROC (%)Sensitivity (%)Specificity (%)Calculated Prevalence (%)
Logistic regression67.0039 (20–61)95 (87–99)13 (7–21)
Bayes Naïve67.0039 (20–61)95 (87–99)13 (7–21)
Decision tree55.2713 (3–34)97 (91–100)5 (2–11)
Boosted Decision tree79.1222 (7–44)99 (93–100)6 (2–12)
Neural Network80.6335 (16–57)95 (87–99)12 (6–20)
SVM76.009 (1–28)99 (93–100)3 (1–8)
Random Forrest71.4023 (15–32)96 (89–99)7 (3–14)
Sample size = 101 PD patients. Actual pRBD prevalence was 23% (15–32%).
Table 4. Odds ratios and performance of ML models, including clinical variables for predicting probable RBD in the “validation” subsample.
Table 4. Odds ratios and performance of ML models, including clinical variables for predicting probable RBD in the “validation” subsample.
Logistic Regression ModelOnly
Genetic
Genetic + ClinicalGenetic + Clinical (LOOCV)
Model variables
GBA_N370S_rs76763715 (Dominant)1.671.201.62
GBA_E365K_rs2230288 (Dominant)3.154.026.45
ACMSD.TMEM163_rs6430538 (Recessive)0.440.57*0.60
MCCC1_rs12637471 (Recessive)3.312.482.34
FAM47E.STBD1_rs6812193 (Recessive)1.911.591.62
SNCA_A53T_rs104893877 (Dominant)6.493.323.65
ANK2.CAMK2D_rs78738012 (Dominant)1.851.762.02
ZNF184_rs9468199 (Dominant)1.852.012.12
CTSB_rs1293298 (Recessive)2.072.252.19
COQ7.SYT17_rs11343 (Recessive)0.400.380.34
ZNF646.KAT8.BCKDK_rs14235 (Dominant)1.531.381.67
Levodopa-Equivalent Daily Dose-1.001.00
MDS-UPDRS I + II + III-1.021.02
Motor Fluctuations-0.891.06
Model parameters
Sample Size10197324
ROC (%)67.082.578.3
Sensitivity (%)39 (20–61)48 (26–70)38 (28–49)
Specificity (%)95 (87–99)97 (91–100)95 (92–98)
Model parameters
Sample Size10197324
ROC (%)67.082.578.3
Sensitivity (%)39 (20–61)48 (26–70)38 (28–49)
Specificity (%)95 (87–99)97 (91–100)95 (92–98)
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Perez-Lloret, S.; Chevalier, G.; Bordet, S.; Barbar, H.; Capani, F.; Udovin, L.; Otero-Losada, M. The Genetic Basis of Probable REM Sleep Behavior Disorder in Parkinson’s Disease. Brain Sci. 2023, 13, 1146. https://doi.org/10.3390/brainsci13081146

AMA Style

Perez-Lloret S, Chevalier G, Bordet S, Barbar H, Capani F, Udovin L, Otero-Losada M. The Genetic Basis of Probable REM Sleep Behavior Disorder in Parkinson’s Disease. Brain Sciences. 2023; 13(8):1146. https://doi.org/10.3390/brainsci13081146

Chicago/Turabian Style

Perez-Lloret, Santiago, Guenson Chevalier, Sofia Bordet, Hanny Barbar, Francisco Capani, Lucas Udovin, and Matilde Otero-Losada. 2023. "The Genetic Basis of Probable REM Sleep Behavior Disorder in Parkinson’s Disease" Brain Sciences 13, no. 8: 1146. https://doi.org/10.3390/brainsci13081146

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