1. Introduction
Neurological disorders (NDs) are increasingly prevalent and represent the leading cause of disability-adjusted life years [
1]. They account for over 6% of the global disease burden [
2], are the second most common cause of death worldwide with nine million deaths annually, and are exacerbated by severe health disparities. For example, approximately 80% of the 50 million people with epilepsy reside in low- and middle-income countries [
3]. NDs can be classified into six major categories that include age-related neurodegenerative disorders (e.g., Alzheimer’s disease, Huntington’s disease, and Parkinson’s disease), mental disorders (e.g., depression, psychosis), neurotoxic disorders (e.g., alcoholism), cerebrovascular disorders (e.g., stroke), neurodevelopmental disorders (e.g., autism), and other complex disorders (e.g., epilepsy). The pathogenesis of CNS disorders is complex and involves a combination of genetic and environmental factors.
There is currently a lack of pre-symptomatic biomarkers to facilitate the early detection of and intervention for NDs. Treating NDs often involves prolonged symptomatic management, carries significant costs, and has an increased risk of adverse drug reactions (ADRs). Moreover, developing effective treatments requires a comprehensive approach that considers each disorder’s unique characteristics as well as drug-drug interactions (DDIs) and the risk of ADRs in polypharmacy. To this, most patients with NDs require multifactorial treatment. However, this carries the risk of ADRs and DDIs due to comorbidity with other conditions such as hypertension, obesity, diabetes and cardiovascular disorders [
4]. ADRs and DDIs are becoming a major health concern worldwide in subjects undergoing treatment [
5,
6] and rank among the top ten leading causes of death and illness in developed countries[
6]. In the United States alone, approximately half a million ADRs are reported annually, with associated direct costs that exceed
$150 billion per year [
7]. Furthermore, ADRs increase hospital admissions, lengthen hospital stays, raise mortality rates and healthcare costs; they may also lead to drug withdrawal from the market [
8]. A focus on precision medicine could improve patient outcomes and reduce the burden of NDs on individuals, families, and society.
Pharmacogenomics, the study of the influence of genetic variability on drug response and toxicity, is a vital component of precision medicine. By providing insight into how patients will respond to specific therapies, pharmacogenomics guides prescription and drug dose decisions. This helps reduce the risk, occurrence and severity of ADRs while optimizing drug efficacy [
9]. Over 50% of drugs currently have a known pharmacogenomic profile that can be used to optimize efficacy and prevent adverse effects [
6]. The pharmacogenomic machinery integrates drug and gene interactions through two pathways: the pharmacogenomic-pharmacokinetic pathway, which occurs during the absorption, distribution, metabolism and excretion (ADME) processes, and the pharmacogenomic-pharmacodynamic pathway, which occurs at the drug-target level [
10]. Proper functioning of these pathways is essential for drug efficacy, and deficiencies or dysfunctions can cause ADRs or toxicity [
11].
Pharmacogenetics accounts for approximately 80% of variability in drug safety and efficacy [
9]. Rare variants make up 50% of the functional variability reported in more than 140 clinically relevant pharmagenes. Over 400 genes, including their encoded enzymes/proteins, influence drug efficacy and safety, and approximately 240 pharmagenes are associated with ADRs [
12]. Pharmacogenetic outcome is influenced by various classes of genes, including pathogenic, mechanistic, metabolic, transporter and pleiotropic genes that comprise the pharmacogenetic machinery. These genes are regulated by epigenetic factors such as DNA methylation, chromatin/histone modifications, and miRNAs [
4,
13]. The enzymes and transporters that are integral to metabolic pathways exhibit a considerable degree of polymorphism. The presence of polymorphisms in the genes that encode these molecular components cause significant variability in interindividual drug responses [
13,
14]. Cytochrome P450 oxidases (CYPs) play a crucial role in regulating drug efficacy and toxicity. CYP1A2, CYP3A4/5, CYP2C9, CYP2C19 and CYP2D6 are among the most important CYPs involved in drug metabolism [
15].
Genomics and epigenetics play a crucial role in the development and progression of neurodegenerative diseases. Epigenetics refers to hereditary changes in phenotype or gene expression that result from chromatin-based mechanisms, which do not involve alterations in the DNA sequence. Both biological and environmental factors modulate epigenetic modifications, which consequently impact gene expression and phenotype [
16]. The accumulation of various epigenetic alterations over the lifespan may contribute to neurodegenerative disorders [
17,
18]. DNA methylation, the most extensively studied epigenetic mark, is a reversible mechanism catalyzed by DNA methyltransferases (DNMTs) that transfer a methyl group from the cofactor SAM (S-adenosyl-l- methionine) to the C5 position of cytosine found in CpG dinucleotides [
19]; this results in the conversion of cytosines to 5-methylcytosines (5mC). DNA methylation changes the stability and accessibility of DNA, which regulates gene expression [
18]. It is most commonly associated with gene silencing [
20] that attracts other silencing elements such as methyl-CpG-binding proteins [
21]. There are three families of DNMT proteins: DNMT1, DNMT2, and DNMT3, and all are expressed in neurons [
22]. DNMT1 is responsible for maintaining methylation patterns after cell division, which allows for the inheritance of methylation marks [
23]. DNMT3a and DNMT3b are responsible for de novo methylation [
24]. Decreased global DNA methylation levels detected in blood samples of patients with neurodegenerative disorders could potentially function as a diagnostic biomarker [
25,
26,
27,
28]. Epigenetic-based therapies may potentially restore 5mC and other epigenetic modifications, providing a novel approach that could delay or reduce disease progression and improve the quality of life of patients.
Our recent study identified two categories of patients: Group A, in which 5mC levels were higher during the follow-up than during the initial visit, and Group B, in which patients had lower or similar 5mC levels during the follow-up than during the initial visit [
29]. Given that pharmacogenetics can account for more than 80% variability in drug pharmacokinetics and pharmacodynamics, we conducted a retrospective study to investigate the influence of genetic factors on global DNA methylation levels in patients with NDs. More specifically, we investigated the influence of metabolic (
CYP1A1,
CYP1A2,
CYP1B1,
CYP2A6,
CYP2B6,
CYP2C9,
CYP2C19,
CYP2D6,
CYP2E1,
CYP3A4,
CYP3A5,
CYP4F2,
CES1,
CHAT,
COMT,
GSTM1,
GSTP1,
GSTT1,
NAT2,
SOD2,
TPMT,
UGT1A1), transporter (
ABCB1,
ABCC2,
ABCG2,
SLC2A2,
SLC2A9,
SLC6A2,
SLC6A3,
SLC6A4,
SLC39A8,
SLCO1B1), and pathogenic (
NBEA,
PTGS2,
APOE) gene variants on global DNA methylation levels, with respect to the therapeutic outcome during the initial and follow-up clinical assessments in both groups of patients.
Our aim in the current study was to provide new insights into the complex interplay between genetic and epigenetic determinants in modulating temporal DNA methylation patterns in patients with NDs to help us better understand disease susceptibility, treatment outcomes, and develop precision medicine strategies.
4. Discussion
DNA methylation has emerged as a promising biomarker for brain-related disorders as it provides insights into gene expression patterns and potential therapeutic targets [
25,
26]. In a recent study, we examined the correlation between hypovitaminosis, psychometric parameters, DNA methylation, and NDs, and found two distinct patient categories based on changes in 5mC levels during clinical follow-up: Group A, which showed increased 5mC levels during the follow-up, and Group B, which exhibited no discernible change in 5mC levels [
29]. In the current study, we investigated, in those two groups of patients, how different pharmacogenetic patient phenotypes, encoded by metabolizing, transporter- and pathogenic genes, affect DNA methylation during clinical follow-up. Our major finding was that variations in genotype-phenotype interactions cause different responses to treatments and are associated with differential changes in 5mC levels over time.
Our first analysis examined the impact of genetic variability in drug metabolizing enzymes on 5mC levels. We focused on different phenotypes of CYP enzymes in the two groups of patients and found that specific SNPs had a significant impact on CYP enzyme activity, leading to changes in phenotypic distribution between the two groups of patients. In particular, there were no significant changes in the phenotypic distribution for some SNPs, while others showed a shift in the percentage of different metabolizing phenotypes.
To identify additional genetic factors that influence drug metabolism and inter-individual differences in patient drug responses, we next analyzed the effects of specific SNPs on CYP and phase II metabolism phenotypes in patient Groups A and B. We found that genetic polymorphisms in CES, CHAT, COMT, GSTM1, GSTP1, GSTT1, NAT2, SOD2, TPMT, and UGT1A1 contributed to the observed differences in drug response between the two groups. Furthermore, there was an increase in normal metabolizers in some genes, whereas others showed a decrease in poor metabolizers and an increase in intermediate metabolizers.
By analyzing transport gene phenotypes, we then assessed whether specific SNPs could modify DNA methylation levels and alter phenotypic responses to treatment. Specifically, we examined the frequencies of normal, deficient, and abnormal response phenotypes for each gene in patient groups A and B. There were significant differences in phenotype distribution between the two patient groups for several SNPs associated with ABCB1, ABCC2, ABCG2, SLC2A2, SLC2A9, SLC6A2, SLC6A3, SLC6A4, SLC39A8, and SLCO1B1. SLCO1B1 SNPs had a strong effect on patient response to treatment, with a marked increase in the frequency of the normal response phenotype in Group B compared to Group A.
Finally, to understand the role of pathogenic gene phenotypes in modifying DNA methylation levels and their impact on disease risk and drug response, we then analyzed the distribution of different genotypes in the two groups of patients and found substantial differences in the frequencies of APOE, NBEA, and PTGS2 genotypes.
CYP enzymes constitute a diverse group of drug-metabolizing enzymes that regulate the metabolism of the majority of xenobiotic substances [
48]. CYPs are responsible for approximately 80% of oxidative metabolism and 50% of drug elimination for currently used drugs [
6]. The normal metabolizer (NM), intermediate metabolizer (IM), poor metabolizer (PM), and ultra-rapid metabolizer (UM) phenotypes, linked to different gene variants, determine drug efficacy and toxicity [
6,
49,
50]. CYP1A2, for example, metabolizes various drugs, including phenacetin, caffeine, clozapine, tacrine, propranolol, and mexiletine [
51]. In particular, a strong correlation exists between coffee consumption and PD among slow metabolizers of caffeine homozygous for
CYP1A2 polymorphisms [
52]. In patients with schizophrenia, clozapine dosage is influenced by CYP1A2 activity, and tacrine metabolism is primarily dependent on the activity of CYP1A2 and CYP3A4 enzymes [
53,
54]. Tacrine, a reversible cholinesterase inhibitor [
55], is a major substrate of CYP1A2 and CYP3A4 [
56]. Propanolol is commonly prescribed for migraine prophylaxis and anxiety treatment [
57,
58]. Our findings suggest that Group B, in which patients did not show an improvement in 5mC levels during follow-up, contained a lower frequency of subjects that were normal metabolizers (40%, compared to 75% in Group A) and a higher frequency of patients that were ultra-rapid metabolizers. CYP1A2 is important for the dosing of several antipsychotics. Ultra-rapid metabolizers are resistant to clozapine treatment, and improved outcomes are achieved by co-administration of the CYP1A2 inhibitor fluvoxamine and by increasing clozapine dosage [
59]. The higher frequency of ultra-rapid metabolizers in Group B patients may therefore explain the poor response to treatment and lack of improvement in 5mC levels during the clinical follow-up.
CYP2E1 is involved in the metabolism of fatty acids, which are abundant in the brain [
60], and in the biotransformation of exogenous compounds [
61]. These compounds include ethanol, nicotine, acetaminophen, acetone, aspartame, chloroform, chlorzoxazone, tetrachloride, and antiepileptic drugs such as phenobarbital. In transgenic (APP/PS1) AD mice, chlorzoxazone is neuroprotective by attenuating neuroinflammation and neurodegeneration [
62]. Our results indicate that in Group B, the frequency of the normal metabolizer phenotype was lower (76% vs. 89%) and the frequency of the intermediate metabolizer phenotype was higher (24% vs. 11%) compared to Group A. Intermediate metabolizers display reduced enzymatic activity and increased side effects because of incomplete drug metabolism compared to normal metabolizers, indicating that a lower dose may be required [
63]. The increase in the frequency of intermediate metabolizers in Group B subjects may contribute to the observed increase in toxicity issues and lack of improvement in 5mC levels during the patient follow-up period.
CYP4F2 is involved in the metabolism of fatty acids and vitamin E [
64]. Vitamin E is associated with decreased DNMT expression [
65]. Our current findings showed that Group B had a higher frequency of the normal metabolizer phenotype (82%) compared to Group A (52%). Moreover, the frequencies of intermediate and poor metabolizers were lower in Group B. This suggests that patients in Group B may be able to metabolize vitamin E more effectively than patents in Group A, which could potentially decrease DNA methylation.
Variants in the
CHAT gene may influence the response to AChEIs [
66]. In our study, the frequency of the intermediate metabolizer phenotype increased from 27% to 48% in Group B. Intermediate metabolizers have reduced enzymatic activity and increased side effects, which could explain the lack of response to treatment and the absence of improvement in 5mC levels in Group B patients. Similarly, COMT analysis showed an increase in the frequency of the intermediate metabolizer phenotype from 47% (Group A) to 68% in Group B. COMT enzyme activity is linked to various psychiatric and neurological disorders [
67]. COMT is involved in Phase II metabolism and transfers a methyl group from S-adenosylmethionine (SAM) to a hydroxyl group on the catechol ring of endogenous and xenobiotic catechol substrates. During COMT-catalyzed methylation, SAM is converted to a competitive inhibitor, S-adenosylhomocysteine (SAH), resulting in a negative feedback loop [
67]. Endogenous substrates of COMT include dopamine, norepinephrine, and epinephrine [
68] and there is a significant association between the
COMT rs4680 SNP and the response to antidepressant treatment [
67].
Although previous studies found no significant correlations between
NAT2 genotypes and AD or PD [
69,
70], our present study found a higher frequency of rapid acetylators in Group B (24%) than in Group A (35%) patients. This increased frequency of rapid acetylators could contribute to the stable levels of 5mC. AtreMorine, a novel compound obtained from the
Vicia faba L. plant using a non-denaturing biotechnological process, increases DNA methylation in patients with PD by providing L-DOPA, a dopamine precursor commonly used to treat PD [
71,
72]. Pharmacogenomic analysis of NAT2 phenotypes in response to AtreMorine show that the increase in DNA methylation is not statistically significant in patients with the fast acetylator phenotype [
72].
Polymorphisms in drug transporter genes affect drug pharmacokinetics and ultimately drug concentration in plasma and target tissues [
73]. Among these transporters, ABCB1 plays a critical role in the brain [
74]. In PD, the frequency of ABCB1 phenotypes is 24.76% for low responders (LR), 32.67% for intermediate responders (IR), and 42.57% for high responders (HR), with no significant differences in response to AtreMorine among the phenotypes [
75]. ABCB1 also transports 42% of antiepileptic drugs and 16% of benzodiazepines [
43]. ABCB1 is a major transporter (55%) of antidepressants [
76] and benzodiazepines (16%); antidepressants are substrates (25%) and inducers (3%) of ABCB1 [
76]. Our findings showed a substantial decrease in the high responder phenotype in Group B, from 60% to 32%. Furthermore, SLC39A8 is involved in the transport of a variety of drugs used to treat anxiety, panic attacks, sleep disorders, agitation, and behavioral anomalies [
43]. In our study, the normal response SLCOB1 phenotype increased from 49% (Group A) to 92% in Group B patients, while only an intermediate response phenotype was observed in Group A.
ApoE is the major carrier of lipids and cholesterol in the CNS [
77]. The expression of the
APOE4 genotype is a significant risk factor for developing late-onset AD [
77]. The severity of synucleinopathies is associated with the
APOE4 variant, independent of concomitant AD severity [
78,
79]. While
APOE4 is not a risk factor for PD, it increases the risk of developing dementia and cognitive decline [
77]. The
APOE genotype also affects the age of onset and severity of stroke, and individuals with the
APOE4 allele exhibit delayed recovery of verbal memory function [
80] and an increased risk of developing stroke-associated dementia [
81]. We and others have shown that
APOE expression decreases in venous blood and plasma samples in AD patients, suggesting its potential as a diagnostic biomarker for the disease [
82,
83].
APOE mRNA expression is also lower in E4 carriers than in individuals with
APOE 2.3 and -
3.3 genotypes [
29]. The incidence of the
APOE4 allele was higher in Group B (20% 3.4, 8% 4.4, and 4% 2.4) than in Group A (16% 3.4 and 3% 4.4), which may explain the lack of improvement in 5mC levels during the follow-up.
In patients with AD, the
NBEA SNP rs17798800 is a potential predictor of response to treatment with AChEIs [
84]. In patients with AD and depression, the observed phenotype frequency of this SNP for AChEI responders (AR), AChEI intermediate responders (DR), and AChEI non-responders (NR) phenotypes are 8.72%, 22.56%, and 68.72%, respectively [
85], which is similar to the phenotype distribution in Group A of our patient cohort. However, in the current study, we observed a different frequency distribution of the NBEA phenotype in Group B patients, with a significant decrease in the normal response frequency (10% AR, 55% DR, and 35% NR). This difference in frequency distribution suggests that the NBEA phenotype may play a role in the changes in 5mC levels observed during the follow-up period.
The present study proposes that analyzing genotypes related to metabolism, transporters, and pathological pharmacogenetics can be used to analyze the evolution of global DNA methylation levels (
Table 1). Other studies have highlighted the correlation between neurodegenerative diseases and decreased levels of sirtuin activity, neurodegenerative gene expression, and global DNA methylation [
25,
26,
82]. Furthermore, DNA methylation has shown potential as a biomarker for monitoring disease outcome in patients with neurological disorders [
29].