Next Article in Journal
Circulating Small EVs miRNAs as Predictors of Pathological Response to Neo-Adjuvant Therapy in Breast Cancer Patients
Next Article in Special Issue
Minor Changes for a Major Impact: A Review of Epigenetic Modifications in Cell-Based Therapies for Stroke
Previous Article in Journal
Mitochondria-Targeting Polymer Micelles in Stepwise Response Releasing Gemcitabine and Destroying the Mitochondria and Nucleus for Combined Antitumor Chemotherapy
Previous Article in Special Issue
Non-Pharmacological Therapeutic Options for the Treatment of Alzheimer’s Disease
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Epigenetic Changes in Prion and Prion-like Neurodegenerative Diseases: Recent Advances, Potential as Biomarkers, and Future Perspectives

by
Adelaida Hernaiz
1,
Janne Markus Toivonen
1,2,
Rosa Bolea
3 and
Inmaculada Martín-Burriel
1,2,3,*
1
Laboratorio de Genética Bioquímica (LAGENBIO), Facultad de Veterinaria, Universidad de Zaragoza, IA2, IIS Aragón, 50013 Zaragoza, Spain
2
Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto Carlos III, 28029 Madrid, Spain
3
Centro de Encefalopatías y Enfermedades Transmisibles Emergentes (CEETE), Facultad de Veterinaria, Universidad de Zaragoza, IA2, IIS Aragón, 50013 Zaragoza, Spain
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(20), 12609; https://doi.org/10.3390/ijms232012609
Submission received: 6 September 2022 / Revised: 9 October 2022 / Accepted: 18 October 2022 / Published: 20 October 2022
(This article belongs to the Special Issue Epigenetics in Neurodegenerative Diseases)

Abstract

:
Prion diseases are transmissible spongiform encephalopathies (TSEs) caused by a conformational conversion of the native cellular prion protein (PrPC) to an abnormal, infectious isoform called PrPSc. Amyotrophic lateral sclerosis, Alzheimer’s, Parkinson’s, and Huntington’s diseases are also known as prion-like diseases because they share common features with prion diseases, including protein misfolding and aggregation, as well as the spread of these misfolded proteins into different brain regions. Increasing evidence proposes the involvement of epigenetic mechanisms, namely DNA methylation, post-translational modifications of histones, and microRNA-mediated post-transcriptional gene regulation in the pathogenesis of prion-like diseases. Little is known about the role of epigenetic modifications in prion diseases, but recent findings also point to a potential regulatory role of epigenetic mechanisms in the pathology of these diseases. This review highlights recent findings on epigenetic modifications in TSEs and prion-like diseases and discusses the potential role of such mechanisms in disease pathology and their use as potential biomarkers.

1. Introduction

Epigenetics is the study of heritable changes in gene activity or function that is not associated with any change in the DNA sequence itself [1]. Three major epigenetic mechanisms have been described: DNA methylation and histone modifications essentially alter the structure of the nearby chromatin of a particular gene or region, whereas non-coding RNAs, especially microRNAs (miRNAs), may regulate expression without the necessity for the physical vicinity for their target genes.
DNA methylation involves the covalent transfer of a methyl group to the C-5 position of the nucleobase cytosine to form 5-methylcytosine. In mammals, DNA methylation may occur at cytosines in any context of the genome [1] and can participate in the regulation of gene expression. Histones are structural proteins that form DNA nucleosomes and are frequently subjected to covalent post-translational modifications. They are involved in the expression and repression of target genes generally via chromatin modification [2]. The most studied histone modifications are acetylation, methylation, and phosphorylation, but also other modifications exist, including citrullination, ubiquitination, ADP-ribosylation, deamination, and proline isomerization [2,3].
MicroRNAs are small non-coding RNAs (21–23 nucleotides in length) that may regulate gene expression at transcriptional, posttranscriptional, or translational levels [4]. Although miRNAs may induce gene expression in some cases by their interaction with gene promoters [5], miRNA-based regulation is thought to rely largely on the repression of target mRNA translation or its sequence-dependent degradation. Several miRNAs display a cell- or tissue-specific expression profile, while others are widely expressed. They can be found in blood and other biofluids as free (mainly protein-associated) complexes or can be contained by extracellular vesicles. Moreover, circulating miRNAs can be detected and measured by highly sensitive and specific methods (e.g., quantitative PCR and next-generation sequencing). All these characteristics have facilitated studies of miRNAs as potential biomarker molecules [6].
The mentioned epigenetic mechanisms are involved in several aspects of brain development as well as in normal aging [7,8]. DNA methylation and histone acetylation are essential in memory acquirement, learning, and acquisition of long-term memories [8,9,10], and miRNAs are crucial for the formation and maturation of synapses and for dendritogenesis in early brain development [11,12,13]. Considering the central role of epigenetics in neural plasticity, it is not surprising that dysregulation of these processes has been associated with different neurodegenerative diseases, either by mediating interactions between genetic and environmental risk factors or by directly interacting with disease-specific pathological factors [7,8]. The neurodegenerative disorders where epigenetics seem to have a key role include Alzheimer’s (AD), Parkinson’s (PD), and Huntington’s (HD) diseases, amyotrophic lateral sclerosis (ALS), prion diseases, stroke, and global ischemia [7,8]. The first four are also known as prion-like diseases because they are proteinopathies that share common pathogenic mechanisms with prion diseases, including the accumulation of misfolded proteins in the central nervous system (CNS) [14,15].
Prion diseases, also referred to as transmissible spongiform encephalopathies (TSEs), are a group of neurodegenerative disorders affecting humans and other animals [16] and are caused by a conformational conversion of the cellular prion protein (PrPC) to an infectious isoform, partially resistant to proteases and prone to form aggregates called PrPSc [17].
Research in yeast has revealed that certain proteins that adopt prion conformation, such as URE3 or PSI+, can be considered as an epigenetic mechanism that can be inherited through mitosis and meiosis. These self-templating conformations of prion proteins interact with nucleic acids (DNA and RNA) and can regulate gene expression through the modification of chromatin remodeling, nucleic acid translation, and replication, providing beneficial phenotypes in stressful conditions [18]. If prions in yeast possess these characteristics, it could be expected that alteration of the prion protein and other proteins with similar characteristics in mammals will also affect epigenetic mechanisms. Increasing evidence suggests involvement of epigenetic mechanisms in the pathogenesis of prion-like diseases and other neurodegenerative disorders. However, current knowledge about the association of epigenetics with prion diseases is scarce.
Given that some of the affected mechanisms may be shared with TSEs and prion-like diseases, this review summarizes the latest findings on epigenetic modifications in prion-like diseases and reports the recently discovered roles of epigenetic mechanisms in TSEs.

2. Epigenetic Changes in Prion-Like Diseases

2.1. DNA Methylation

2.1.1. DNA Methylation Profiles

Altered DNA methylation patterns have been associated with the neuropathology of prion-like diseases. Several studies have identified sets of genes containing differentially methylated regions (DMRs) and differentially methylated positions (DMPs) between patients and healthy controls, primarily in peripheral blood [19,20,21,22,23,24,25,26] and brain tissue [27,28,29,30,31,32,33,34,35] and to a lesser extent in saliva [23] and using in vitro models [36,37]. Table 1 summarizes global and gene-specific DNA methylation changes reported in these diseases.
When comparing the DMRs and DMPs identified in each prion-like disease (Supplementary Table S1) using the InteractiVenn software [38], no common DMRs and DMPs were found between all four diseases, but some regions and positions matched between two or three of these diseases (Figure 1). Regarding the DMRs, the disease pairs that shared the highest number of common regions were first PD and HD, second PD and ALS, and third PD and AD (Figure 1a). On the other hand, PD and AD are the only diseases that share a relatively high number of DMPs (Figure 1b). Although these comparisons between the DMRs and DMPs identified in prion-like diseases allow an overview of common differentially methylated genes shared between some of these diseases, there are limitations that could affect the number of regions and positions identified in each disease and must be taken into account: (1) the compared studies were performed in different tissues and body fluids which could lead to different methylation profiles inherent to each tissue; (2) the number of studies developed in each disease is different, being AD and PD the most studied ones; and (3) the methodology used in the different works varies. Most studies were performed using the 450 K methylation array, and whole genome bisulfite sequencing was used only in one study of PD [37]. This methodology detects DMPs in non-annotated sequences such as lincRNAs, pseudogenes, and antisense or unknown miRNAs, which explains in part why the number of DMPs and DMRs in PD is substantially higher than in other diseases.
Studies on the DNA methylation process have concentrated on different areas of the CNS depending on their relevance with the pathogenesis of each disease. Epigenome-wide studies on AD patients have identified DMRs in the superior temporal gyrus, a region that displays a marked gene dysregulation in AD [27,28]. Most of these DMRs were found to be in gene promoters and associated with AD pathology. Significant DNA methylation changes are also found in PD-affected brain areas, namely the dorsal motor nucleus of the vagus, substantia nigra, and cingulate gyrus [32]. In HD, DNA methylation studies performed so far have shown disparate results. A genome-wide DNA methylation profile of cortex tissues from HD patients suggests that DNA methylation may have a minimal association with HD status but could be correlated with the age of disease onset and contribute to the tissue-specific expression patterns of huntingtin (HTT) [39]. In contrast, using the same methodology but analyzing multiple CNS regions, another study identified 11 co-methylation modules associated with HD status in cortical regions and observed an accelerated epigenetic age in specific brain regions (frontal lobe, parietal lobe, and cingulate gyrus) of HD patients [35], measured by combining DNA methylation levels of known CpGs [40].
Genome-wide studies have detected DNA modifications in specific genes that have been analyzed in detail to elucidate their role in neurodegeneration. Epigenetic control of enhancers seems to be involved in AD as loss of CpH (most frequently at CpA sites) methylation of enhancers, which is normal in aging neurons but is accelerated and occurs early in AD neurons. This modification seems to prompt the reactivation of cell cycle and neurogenesis pathways and, in the case of the enhancer of DSCAML1 that codes for Down Syndrome cell adhesion molecule-like protein 1, precedes the onset of neurofibrillary tangle pathology [41]. Hypomethylation of DSCAML1 gene enhancer is associated with an upregulation of BACE1 (beta-secretase 1) transcripts and an increase in amyloid plaques, neurofibrillary tangles, and cognitive decline [41]. These results support an early involvement of epigenetic changes in AD.
In the same disease, a protective role has been suggested for PM20D1 (peptidase M20 domain containing 1). DNA methylation and RNA expression of this gene are associated with AD, and its overexpression in cells is neuroprotective against AD stressors [42].
Methylation changes in other genes have been related to prion-like disease pathogenesis, although its role is still unknown. In PD, patients show hypomethylation of the promoter region of SNCA, the gene encoding α-synuclein protein [43]. Moreover, several ALS-related genes (DENND11, COL15A1, TARDBP, RANGAP1, and IGHMBP2) and DNA repair genes (OGG1, APEX1, PNKP, and APTX) are differentially methylated in ALS patients [44,45,46]. Differential DNA methylation has also been observed in specific genes in HD, namely HES4, a transcription factor involved in neural stem cell regeneration, and BDNF, encoding brain-derived neurotrophic factor [47,48]. HES4 promoter hypermethylation is associated with reduced expression of the gene and with striatal degeneration and age of onset of HD patients. Consistently, inhibition of HES4 by shRNA increases mutant HTT aggregates in a cell model of the disease. BDNF promoter hypermethylation in the blood of HD patients did not correlate with motor or cognitive status but may represent a biomarker for HD-associated psychiatric symptoms.
Most of the reported studies reveal that changes in DNA methylation can be associated with pathogenic processes of these neurodegenerative diseases, but there does not appear to be a universal mechanism relating DNA methylation and protein aggregates. Studies performed in human patients are normally carried out at the late stages of the disease when it is difficult to differentiate if changes in DNA methylation are a cause or a consequence of neurodegeneration. Analysis of this epigenetic process in animal or cellular models could possibly allow us to elucidate this question.
A limitation of most of these studies is the technology used for the determination of DNA methylation. Methylation arrays or NGS-sequencing-based methods did not differentiate between methyl Cytosines (mC) and their oxidized product hydroxymethyl Cytosines (hmC), which has regulatory functions and could be an epigenetic mark in its own right [49]. Antibody-based techniques have already been used in these diseases to specifically detect 5mC and 5hmC methylation forms [50,51]. However, these techniques cannot identify specific genes of interest. Third-generation sequencers [52] could help elucidate which epigenetic mark is involved in a gene-specific regulation.

2.1.2. Biomarkers Based on DNA Methylation

DNA methylation as a potential biomarker has been investigated in easily accessible tissues at both genomic and gene-specific levels. Both sporadic (SALS) and familial ALS (FALS) patients show increased global 5-methylcytosine levels in blood DNA [53]. At the gene level, hypermethylation of the promoter for C9orf72, a gene responsible for the majority of the FALS cases (as well as for those of frontotemporal dementia), correlates with its reduced mRNA expression levels in a clinical cohort of C9orf72 pathological expansion carriers [54]. A significant association between DNA methylation age-acceleration, disease duration, and age of onset has also been observed in C9orf72 carriers [55]. The second most common cause of FALS is mutations in superoxide dismutase 1 (SOD1), with nearly 200 mutations described, some of which show incomplete penetrance and great phenotypic variability. ALS patients carrying not fully penetrant SOD1 mutations display an increase in global DNA methylation, and DNA methylation levels correlate positively with disease duration [56]. However, since the promoters of four major ALS genes (C9orf72, SOD1, TARDBP, and FUS) were not methylated in the study subjects, it was concluded that the increased methylation is likely to occur in other gene regions. In this study, no repeat expansion was observed in C9orf72, which supports the previous finding [51] that gene-specific methylation in the C9orf72 locus is dependent on this pathological feature.
In contrast, DNA hypomethylation seems to be a key mark in PD patients, finding several hypomethylated and upregulated genes in blood and saliva samples associated with systemic immune response pathways and mitochondrial dysfunction [21,23]. In addition, in leukocytes from PD patients, hypomethylation of NPAS2 (neuronal PAS domain protein 2) [57] and DRD2 (dopamine receptor D2) [57] has been proposed as a novel biomarker for PD.
In blood from AD patients, B3GALT4 (beta-1,3-galactosyltransferase 4), a gene associated with AD onset and progression, and PTGR3 (prostaglandin reductase 3), associated with AD risk, are hypomethylated and correlate with memory performance and cerebrospinal fluid (CSF) levels of Aβ and tau [58]. Moreover, hypomethylation of BIN1 (bridging integrator 1), a gene associated with AD pathogenesis, and hypermethylation of estrogen receptor α (ESR1) gene promoter, which is related to impaired cognitive function and quality of life of AD patients, have been also reported [59,60]. Another potential biomarker of this disease could be the hypomethylation in TOMM40 (translocase of outer mitochondrial membrane 40) and APOE (apolipoprotein E) gene promoters observed in the hippocampus, cerebellum, and peripheral blood of AD patients, which correlated with increasing APOE and decreasing TOMM40 expression [61]. Besides these promising results, very few have been subjected to clinical trials. Only methylation levels on COASY (Coenzyme A synthase) and SPINT1 (Serine peptidase inhibitor Kunitz type 1) promoter regions have been considered convenient and useful biomarkers for AD [62].
In HD, DNA methylation status in peripheral blood does not seem to be affected as, after a microarray study of blood samples, no distinctive patterns were observed in the covered CpG sites and their associated genes [25].

2.1.3. In Vitro Studies

Cellular models are particularly useful for analyzing the effect of DNA methylation and investigating the role of methylation in candidate genes and potential treatments. DNA methylation changes in prion-like diseases have been investigated in vitro using cell models. A whole-genome bisulfite sequencing study using induced pluripotent stem cell (iPSC)-derived dopaminergic neurons from sporadic PD and monogenic LRRK2 (Leucine-rich repeat kinase 2)-associated PD patients revealed global DNA hypermethylation associated with disease [37], conversely to the hypomethylation observed in blood and saliva. In ALS, a study that evaluated the methylation status of human embryonic stem cells (hESCs) and iPSCs both carrying the C9orf72 mutation showed that hESCs were completely unmethylated at the C9orf72 repeats, whereas iPSCs were hypermethylated. This methylation status remained unchanged after the differentiation of hESCs and iPSCs into neural precursors. The hypermethylation observed in the C9orf72 repeats of iPSCs was proposed as a possible neuroprotective mechanism attenuating the accumulation of potentially toxic repeat-containing mRNAs in neurons, given that hESCs presented a more severe phenotype than iPSCs [63].
Hypomethylation of the SNCA gene has been described in early onset PD patients [43], and lowered methylation status is likely to increase SNCA expression, contributing to the accumulation of α-synuclein in this disease. With the purpose of maintaining normal physiological levels of α-synuclein, Kantor et al. [64] experimentally methylated the SNCA gene using a system based on CRISPR-deactivated Cas9 fused with the catalytic domain of DNMT3A (DNA methyltransferase 3 alpha). This technique was applied to iPSC-derived dopaminergic neurons from a PD patient resulting in hypermethylation of the SNCA gene, downregulation of the SNCA expression, and a reversion of disease-related phenotypic perturbations [65]. The same hypermethylation-based approach has been recently used to manipulate the levels of amyloid-beta (Aβ) precursor (APP) in cultured neurons from AD mice, resulting in decreased Aβ peptide levels, decreased Aβ42/40 ratio, and increased cell survival [66]. Importantly, further studies in vivo indicated that lentiviral injection of dCas9-Dnmt3a in mouse brain induces efficient DNA methylation editing, decreases the levels of APP, and improves the cognitive defects associated with this AD model.

2.1.4. Mitochondrial DNA Methylation

Finally, in addition to nuclear DNA, mitochondrial DNA (mtDNA) methylation has also been observed in humans and animal models. In ALS, SOD1 mutation carriers display increased levels of mtDNA and demethylation of the mitochondrial D-loop, a noncoding region critical for both mtDNA replication and transcription. This could represent an attempt to compensate for the disease-associated loss of mitochondrial function by mtDNA upregulation in carriers of ALS-linked SOD1 mutations [67]. While D-loop is also hypomethylated in the hippocampus of a mouse model of AD (APP/PS1), this is associated with decreased mtDNA copy number [68]. Thus, while altered methylation of mtDNA suggests a possible role for this epigenetic mechanism in ALS and AD, the fact that the outcome in the level of mtDNA is opposite in the two cases warrants further studies on the topic.
Table 1. Global and gene-specific DNA methylation in prion-like diseases.
Table 1. Global and gene-specific DNA methylation in prion-like diseases.
Disease Species/Model Tissue Type Methylation Finding References
Alzheimer’s
disease
HumanBrain, peripheral bloodMethylation profiles of AD patients. Identification of differentially methylated positions (DMPs)[19,20,30,31]
HumanBrainMethylation profile of AD patients. Identification of differentially methylated regions (DMRs)[27]
HumanSuperior temporal gyrusHypermethylated DMRs[28,29]
APP/PS1 miceHippocampusChanges in mitochondrial DNA methylation[68]
HumanNeuronsHypomethylated enhancers in DSCAML1 gene[41]
HumanHippocampus, cerebellum, peripheral bloodHypomethylation in TOMM40 and APOE gene promoters[61]
HumanFrontal cortexMethylation of PM20D1 gene[42]
HumanPeripheral bloodHypomethylation of B3GALT4 and ZADH2 genes[57]
HumanPeripheral bloodHypomethylation of BIN1 gene[58]
HumanPeripheral bloodHypermethylation of ERα gene promoter[59]
Parkinson’s
disease
HumanPeripheral bloodIdentification of DMRs between PD patients and healthy controls[22]
HumanBrainIdentification of DMRs in PD-affected brain areas[32]
HumaniPSC-derived dopaminergic neuronsGlobal DNA hypermethylation changes[36,37]
HumanPeripheral blood, salivaGlobal DNA hypomethylation changes[21,23]
HumanPeripheral blood, iPSC-derived dopaminergic neuronsHypomethylation of SNCA gene promoter. Reversion of disease symptoms via CRISPR/Cas9-mediated SNCA hypermethylation[43,65]
HumanLeukocytesHypomethylation of NPAS2 and DRD2 genes[60,63]
Amyotrophic lateral
sclerosis
HumanPeripheral bloodMethylation profile of ALS patients. Identification of DMPs[24]
HumanBrainMethylation profile of ALS patients. Identification of DMRs[33]
HumanPeripheral bloodIncreased global 5-methylcytosines levels in sALS and FALS[53]
HumanPeripheral bloodHypermethylation of the C9orf72 promoter and association of DNA methylation age-acceleration with disease duration and age of onset in C9orf72 expansion carriers[54,55]
HumanPeripheral bloodIncrease in global DNA methylation and demethylation of the mitochondrial D-loop region in SOD1 mutation carriers[56,67]
HumanhESCs, iPSCshESCs unmethylated and iPSCs hypermethylated at the C9orf72 repeats[64]
HumanBrain, motor neuronsDifferential methylation of KIAA1147, IGHMBP2, COL15A1, TARDBP, RANGAP1, IGHMBP2, OGG1, APEX1, PNKP, and APTX[44,45,46]
Huntington’s diseaseYAC128 miceBrainMethylation profile. Identification of DMRs[34]
HumanPeripheral bloodMethylation profile of HD patients. Identification of DMPs[26]
HumanBrainMinimal association of DNA methylation with HD status[39]
HumanPeripheral bloodNo significant changes between patients and controls[25]
HumanBrain11 co-methylation modules associated with HD status[35]
HumanBrainHES4 promoter hypermethylation[47]
HumanPlasma, salivaBDNF promoter hypermethylation[48]

2.2. Histone Modifications

The most studied histone modification in prion-like diseases is acetylation. Global levels of acetylated histones and differential expression of the enzymes in charge of their acetylation or deacetylation have been related to the pathology of these diseases. Moreover, pharmacological modulation of these enzymes could ultimately result in a potential treatment strategy. Table 2 summarizes the main results in this subject.
Widespread acetylome variation has been observed in different brain areas of AD patients, and this epigenetic modification can be induced by pathological tau [69,70]. Acetylation of a lysine residue in histone 3 (H3K27) varies in the vicinity of several known AD risk genes (APP, CR1, MAPT, PSEN1, PSEN2, and TOMM40) and is robustly associated with the disease [69]. Similarly, altered histone acetylation has been linked to PD-associated neurodegeneration. PD neurotoxins specifically increase histone acetylation through an autophagy-mediated reduction of histone deacetylases (HDACs) in PD patients’ dopaminergic neurons [71].
The modulation of epigenetic enzymes involved in acetylation, mainly HDACs, also has a role in the development and progression of prion-like diseases. HDACs can be divided into four subtypes, classes I, II, and IV being classical HDACs and class III consisting of NAD+-dependent silent information regulator 2 family members (sirtuins). HDAC activities are unbalanced in fibroblasts from PD patients, which is associated with impaired mitophagy and increased cell death [72].
In addition, changes in expression levels of particular HDACs have been described, but their modifications are not always in accordance with studies. Modifications can be different depending on the disease and CNS cell populations. HDAC1 and HDAC2 levels are strongly decreased in the frontal cortex of AD patients, and HDAC1 is also reduced in the hippocampus [73]. HDAC2 downregulation seems to contribute to cholinergic nucleus basalis of Meynert neuronal dysfunction, neurofibrillary tangles pathology, and cognitive decline during the clinical progression of AD [74]. On the contrary, HDAC2 is upregulated in the microglia from the substantia nigra of PD patients [75]. Additionally, nuclear accumulation of HDAC4, which is normally localized at the cytoplasm, seems to promote neuronal apoptosis in PD-affected dopaminergic neurons [76].
The levels of class II HDACs (4, 5, and 6) are increased in the skeletal muscle of SOD1-ALS mice with severe neuromuscular impairment [77]. Although this increase was not observed in motor neurons, Class II HDACs could also contribute to motor neuron degeneration as their pharmacological inhibition is able to restore the expression and function of glutamate transporter EAAT2 in the spinal cord of ALS [78]. However, HDAC4 expression is decreased in the skeletal muscle of ALS patients [79], and the skeletal muscle-specific ablation of HDAC4 is sufficient to induce an earlier onset of the disease, a decrease in neuromuscular junctions’ size, and muscle denervation and atrophy in SOD1-ALS mice [80]. Finally, an in vivo brain assessment of HDAC alterations by positron emission tomography showed no significant differences in HDAC expression levels between ALS patients and healthy controls [81], suggesting that HDAC alterations may have a more profound effect on the disease in peripheral tissues. Given that the skeletal muscle function is compromised in ALS, the implication that HDAC4 could be protective in ALS skeletal muscle has questioned the use of broad-range HDAC inhibitors as a strategy for ALS treatment [82]. However, research on the role of HDACs in different tissues warrants further investigation because the discovery of highly selective inhibitors for HDACs could eliminate the potential negative effects of more broad-range targeting in the future.
In AD patient-derived neurons and triple transgenic (3xTg-AD, carrying mutated forms of APP, PSEN1, and MAPT/tau) mouse model, inhibition of HDAC3 (Class I) decreases pathological tau phosphorylation and acetylation, reduces Aβ protein expression, increases Aβ degradation, improves learning and memory and normalizes several AD-related genes [83]. Importantly, HDAC6 seems to influence tau phosphorylation, autophagic flux, and tubulin acetylation [84], and its inhibition stimulates pathological tau degradation in ADLP APT mice (carrying six mutations in APP, PSEN1, and MAPT/tau) and AD patient-derived brain organoids [85]. The inhibition of HDAC6 has also been proposed as a therapeutic strategy for PD, as this enzyme contributes to oxidative injury and dopaminergic neurotoxicity through mediating deacetylation of peroxiredoxins 1 and 2 in oxidopamine-induced PD mice [86]. In the PD mouse model expressing mutated LRRK2 (R1441G), HDAC inhibition by valproic acid has a neuroprotective effect through modulation of neuroinflammation and improvement of PD-like symptoms [87]. Inhibition of histone sirtuin-2 deacetylase (SIRT2) also has therapeutic effects protecting degenerating dopaminergic neurons, reducing microglia activation, and facilitating the trafficking and clearance of misfolded proteins [88,89].
This therapeutic approach of HDAC inhibition has also been investigated in ALS models. HDAC inhibition in spinal cord-dorsal root ganglion cultures enables the heat shock response, which manages a load of aberrant proteins in a stress-dependent manner in cultured spinal motor neurons and also rescues the DNA repair response in iPSC-derived motor neurons carrying the FUS mutation [90]. Similarly, global histone hypoacetylation was observed in a FUS murine model. The restoration of histone acetylation levels in these mice by HDAC inhibition ameliorated the disease phenotype and significantly extended their lifespan [91]. In addition, HDAC6 inhibition restored axonal transport defects and mitochondrial and endoplasmic reticulum vesicle transport defects in ALS patient-derived motor neurons by increasing the α-tubulin acetylation level [92,93].
In HD rats and mouse models, HDAC inhibition produces a variety of neuroprotective beneficial effects, including partial reversal of behavioral symptoms, reversion of aberrant neuronal differentiation [94], prevention of striatal neuronal atrophy, improvement of motor performance [95], amelioration of disease phenotypes in a transgenerational manner [96] and reestablishment of pyruvate dehydrogenase activity improving mitochondrial function and bioenergetics [97]. Moreover, a multiomic study has proposed that the positive effect of HDAC4 knockdown in rescuing synaptic function in HD mice could be a consequence of synaptic vesicle trafficking regulation, and HDAC4 could interact with htt via association with htt-interacting proteins [98]. Other HDACs have also been proposed as therapeutic targets in HD. Striatal HDAC2 levels are reduced in the YAC128 HD mouse model subjected to dietary restriction, which could contribute to improving the disease phenotype [99]. In addition, inhibition of HDAC3 improves motor deficits, suppresses striatal CAG repeat expansions, and reduces the accumulation of oligomeric forms of mutant htt in HD transgenic mice [100,101]. In contrast, genetic deletion of HDAC6 exacerbates social impairments and hypolocomotion in HD R6/1 mice [102].
Although there are fewer works analyzing this epigenetic mechanism compared to DNA methylation or microRNAs, the results obtained seem to be promising, not as a source of biomarkers but as a possible target for therapies.
Other histone modifications have been studied, but the number of these studies is even lower. A genome-wide study in human brains has identified differential enrichment of trimethylated lysine 4 of histone 3 (H3K4me3) mark between HD and control samples [103]. Interestingly, in a Drosophila melanogaster HD model, the activity reduction of the H3K27-specific demethylase, Utx, ameliorated neurodegeneration and diminished htt aggregation [104]. Conversely, in this same model, histone methyltransferase dSETDB1/ESET was identified as a mediator of mutant htt-induced degeneration [105].
Table 2. Histone post-translational modifications in prion-like diseases.
Table 2. Histone post-translational modifications in prion-like diseases.
Disease Species/Model Tissue Type Main Finding References
Alzheimer’s diseaseHumanBrainWidespread acetylomic variation associated with AD, possibly induced by pathological tau[69,70]
HumanFrontal cortex, hippocampusDecreased levels of HDAC1[73]
HumanFrontal cortexDecreased levels of HDAC2 contributing to neuronal dysfunction, neurofibrillary tangles pathology, and cognitive decline[73,74]
3xTg-AD mice, HumanHuman iPSC-derived neuronsHDAC3 inhibition decreases pathological tau phosphorylation and acetylation[75]
ADLPAPT mice, HumanHuman-derived brain organoidsHDAC6 inhibition stimulates pathological tau degradation[77]
Parkinson’s diseaseHumanDopaminergic neuronsPD neurotoxins increase histone acetylation through an autophagy-mediated HDACs reduction mechanism[71]
HumanFibroblastsImbalance between total HATs and HDACs activities[72]
LRRK2 R1441G miceBrainHDAC inhibition has a neuroprotective effect through modulation of neuroinflammation and improvement of PD-like behaviors[79]
Sirt2 −/− C57-BL6 mice, HumanBrainSirtuin-2 deacetylase inhibition protects degenerating dopaminergic neurons, reduces microglial activation, and facilitates the trafficking and clearance of misfolded proteins[80,81]
HumanMicroglia from the substantia nigraUpregulation of HDAC2[83]
E13-14 miceDopaminergic neuronsHDAC4 accumulation promotes neuronal apoptosis[84]
C57-BL6 miceBrainHDAC6 could contribute to oxidative injury[86]
Amyotrophic lateral sclerosisSOD1-ALS miceSkeletal muscleIncrease in class II HDACs (4, 5, and 6) involved in modulating the expression and function of glutamate transporter[85,86]
HumaniPSC-derived motor neuronsHDAC inhibition rescues the DNA repair response[90]
Tg FUS +/+ miceSpinal cordGlobal histone hypoacetylation. Restoration of histone acetylation ameliorates the disease phenotype[91]
HumaniPSC-derived motor neuronsHDAC6 inhibition restores axonal transport defects and mitochondrial and endoplasmic reticulum vesicle transport defects[92,93]
HumanBrainNo significant differences in HDAC expression levels between patients and controls[97]
SOD1-ALS mice, HumanSkeletal muscleDecreased expression of HDAC4 associated with an earlier onset of the disease[87,88]
Huntington’s disease tgHD rats, BACHD, R6/2, YAC128 miceBrainHDAC inhibition produces neuroprotective beneficial effects[94,95,96,97]
HttQ20, HttQ140 miceBrainHDAC4 regulates synaptic vesicle trafficking and interacts with htt [98]
YAC128 miceBrainReduction of HDAC2 could contribute to improving the disease phenotype[99]
N171-82Q, HdhQ111 knock-in miceBrainHDAC3 inhibition improves motor deficits, suppresses striatal CAG repeat expansions, and reduces accumulation of mutant huntingtin oligomeric forms[100,101]
R6/1 miceIn vivo assessmentHDAC6 deletion exacerbates social impairments and hypolocomotion[102]
HumanBrainDifferentially enrichment of H3K4me3 mark[103]
Drosophila melanogasterBrain, eyeActivity reduction of Utx ameliorates neurodegeneration and diminishes htt aggregation. dSETDB1/ESET might be a mediator of mutant htt-induced degeneration[104,105]

2.3. MicroRNAs

A large number of human proteins containing prion-like domains are RNA or DNA-binding proteins [106]. The first protein with these motifs associated with neurodegeneration was TDP-43 (trans-activation response element DNA-binding protein 43) [107,108], which is the major pathological protein in sporadic ALS. This protein plays a role in many RNA-related functions, including microRNA biogenesis. Afterward, many other proteins related to neurodegenerative diseases have been discovered that play a role in RNA metabolism, and amyloids coaggregate with endogenous nucleic acids [109,110]. Although different RNA species may be altered in prion-like diseases, we focus on the role of microRNA in these pathologies. These biomolecules have been widely studied in neurodegenerative diseases. Changes in miRNA expression profiles may serve as biomarkers, and these molecules seem to play important roles in the neuropathology of these diseases.

2.3.1. MicroRNA Profiles

Several studies have measured miRNA expression profiles in tissues and body fluids from patients and in different in vitro and animal models of prion-like diseases. Sets of microRNAs altered in each prion-like disease seem to be largely different, as summarized in Table 3.
In AD patients, miRNA profiles have been analyzed in brain [111,112,113,114], blood [115,116], serum [117,118], CSF [119] and extracellular vesicles (EVs) [120,121,122]. Brain and EVs miRNA profiles have as well been characterized in AD mouse models [123,124,125]. Comparing the different profiles, only one miRNA, miR-324-3p, is commonly downregulated in the brain, body fluids, and EVs of AD patients, but its expression is not altered in AD mouse models. MiRNA profiles have been also characterized in brain [126,127,128,129], gut [130], blood [128], plasma [131,132], serum [133], CSF [134,135], saliva [136], and EVs [131,137] of PD patients; in brain of a PD mouse model [135]; in blood of a PD rat model [138]; and in a PD in vitro model [139]. Although no common miRNAs are found when comparing all the above-mentioned PD profiles, there are common miRNAs between some tissues and body fluids. MicroRNA miR-451a is upregulated in the brain, gut, and CSF of PD patients and also in the brains of PD mice, and miR-19b-3p is downregulated in the brain, blood, plasma, and saliva of PD patients.
In ALS patients, microRNAs have been studied in brain [140,141], skeletal muscle [142,143], blood [143,144,145,146], plasma [147], serum [148,149,150], CSF [151] and EVs [152,153,154,155]. Distinctive ALS miRNA profiles are also present in mouse skeletal muscle [156] and serum [157] and in in vitro models [154,158]. Three miRNAs, miR-125a-3p, miR-193a-5p, and miR-455-3p, are commonly downregulated in the brain, skeletal muscle, blood, and serum from ALS patients, and the expression of miR-125a-5p, which is downregulated in the brain and skeletal muscle of ALS patients, is also reduced in ALS mouse models and in vitro models. A smaller number of studies have explored the miRNA profiles in HD. These profiles are found in the brain [159,160,161], plasma [162], and CSF [163] of HD patients and in the brain of HD mouse models [164,165,166,167]. No common miRNAs are found between the profiles performed in HD patients, but there are two miRNAs, miR-10b-5p and miR-10a-5p, that are upregulated in the brain of both HD patients and mouse models.
When comparing the human miRNA profiles of the four prion-like diseases, there are two miRNAs, miR-144-3p and miR-22-5p, that appear upregulated in all four pathologies (Figure 2a). The β-amyloid precursor protein (APP) has been identified as a target gene of miR-144-3p, which seems to have a role in mitochondrial function maintenance [168], and a potential neuroprotective role has been predicted for miR-22 related to the regulation of targets implicated in HD such as HDAC4 [169]. Further analyses are necessary to verify if these two microRNAs display important roles in misfolded protein-related diseases.
Conversely, although there are miRNAs similarly downregulated between two or three prion-like diseases, no common downregulated miRNAs are found between the four diseases (Figure 2b). The existence of differently regulated microRNA in different diseases can be used as a source of biomarkers for differential diagnosis. Supplementary Table S2 lists common upregulated and downregulated miRNAs for combinations of different prion-like diseases and the datasets used to generate the Venn diagrams.

2.3.2. The Role of microRNAs in Prion-Like Diseases

In addition to miRNA profiles that could reveal potential disease biomarkers, some studies have shed light on the functions or possible roles of different miRNAs in prion-like diseases.
In AD pathology, several miRNAs seem to have neuroprotective roles attenuating Aβ accumulation and its associated neurotoxicity. In particular, the following miRNAs have been associated with this neuroprotective function: miR-193a-3p [170] and miR-133b [171] in human serum, miR-335-5p [172] and miR-361-3p [173] in the human brain, miR-200a-3p [174] in plasma from AD patients, miR-107 [175] in human neuroblast cell lines, and miR-340 [176] in the hippocampus from senescence-accelerated (SAMP8) mice that show increased oxidative damage associated with APP overproduction. Moreover, in rat hippocampus, miR-134-5p [177] is involved in rescuing AD synaptic plasticity deficit, and miR-124 [178] and miR-200a-3p [174] are involved in alleviating tau pathology in murine and in vitro models, respectively. On the other hand, miR-34c [179] and miR-124 [180] mediate synaptic and memory deficits in AD. Other miRNAs recently described to likely participate in AD risk and progression are miR-146a, miR-181a, detected in blood from AD patients, and miR-142-3p [181,182] in the human brain. All aforementioned miRNAs have been proposed as potential diagnostic biomarkers and/or therapeutic targets for AD pathology.
Dysregulation of miRNAs is also implicated in PD pathogenesis. The upregulation of several miRNAs, including miR-150 [183] in human serum, miR-let-7a [184], and miR-190 [185] in C57BL/6 mouse model and miR-135b [186] in in vitro models, ameliorates PD-associated neuroinflammation. By inhibiting SP1, a transcription factor expressed in the brain, miR-375 decreases dopaminergic neurons’ damage, reduces oxidative stress, and diminishes inflammation in PD, and miR-29c also attenuates dopaminergic neuron loss, neuroinflammatory response, and α-synuclein accumulation [187,188]. Furthermore, in a PD rat model, miR-3557 and miR-324 seem to be involved in delaying PD neurodegeneration [189], and overexpression of miR-410 in a PD cellular model appears to exert neuroprotective effects against apoptosis and reactive oxygen species production [190]. In contrast, overexpression of miR-326 has been described to promote autophagy of dopaminergic neurons, and miR-195 downregulation might induce microglia-mediated neuroinflammation activation [191,192]. On the other hand, miR-7 seems to be involved in regulating BDNF expression in the early stages of PD, and miR-376a could also be implicated in PD pathogenesis, possibly by regulating the expression of mitochondrial-related genes [193,194].
Several studies also report miRNA changes in ALS, suggesting that these molecules could play a role in the development and progression of the disease. In serum from ALS patients, downregulation of miR-335-5p may enhance mitophagy, autophagy, and apoptosis pathways [195]. Interestingly, in the spinal cord of an ALS mouse model, downregulated miR-375-3p appears to control various target structures that intervene at different sites of the apoptosis pathway [196]. In contrast, in the cerebellum of an ALS mouse model, increased miR-29b-3p seems to downregulate proapoptotic factors, leading to neuroprotection [197]. Regarding to axon degeneration, several miRNAs, namely miR-126-5p [198], miR-494-3p [199] and miR-1825 [200], might facilitate this pathological feature in ALS. On the other hand, upregulated miR-338-3p is responsible for decreased glycogenolysis and subsequent glycogen accumulation within the spinal cord of SOD1-ALS mice [201], and miR-105 and miR-9 seem to potentially contribute to the pathogenesis of intermediate filament inclusions in ALS [202]. Furthermore, it has been described that extracellular miR-218 released from dying motor neurons in ALS can be taken up by neighboring astrocytes and negatively affect astrocyte function [203]. In the skeletal muscle of FALS patients, it has been reported an upregulation of miR-206, involved in the neuromuscular junction, regeneration, and muscle atrophy, and also an increase in inflammatory miRNAs (miR-27a, miR-221, miR-155) [79]. miR-206, a microRNA that was consistently altered during the course of the disease in the skeletal muscle of the SOD1-G93A ALS mouse model, is also increased in serum from ALS patients [155]. Mechanistically, it has been suggested that increased miR-206 is an attempt to promote maintenance and/or repair of neuromuscular junctions by targeting and inhibiting HDAC4, which leads to a fibroblast growth factor (FGF)-stimulated reinnervation [204]. Finally, reduced expression of the miR-17~92 cluster has been associated with the vulnerability of limb-innervating lateral motor column motor neurons to ALS-related degeneration [205].
In HD, a consistent association between expression profiles of CSF-miRNAs and the earliest prodromal stages of the disease has been reported [162]. In contrast, a study in a knock-in mouse model of HD (Hdh mice) suggests that miRNA regulation may have a limited global role in responding to HD in the striatum and cortex of these mice [206]. A decrease in miR-132 has been observed in the brain of another model of HD (HD R6/2 mice), and restoration of miR-132 deficiency seems to confer amelioration in motor function and lifespan of these mice [207]. In neural progenitors and differentiated neural cells of a transgenic HD nonhuman primate model and in HD murine primary neurons, miR-196a has shown neuroprotective effects, including improvement of cell survival and mitochondrial functions, reduction of cytotoxicity and apoptosis and enhancement of neuronal morphology and differentiation [208,209]. Regarding htt, miR-27a has been reported to reduce mutant htt aggregation in R6/2-derived neuronal stem cells [210]. Interestingly, artificial miRNAs have also been used to successfully reduce mutant htt levels in a transgenic HD sheep model and in a humanized Hu128/21 HD mouse model [211,212].
As reported, the role of microRNAs in these neurodegenerative diseases is better known than the ones of the other epigenetic mechanisms. We have the tools to analyze the effect of overexpressing or repressing the expression of these molecules using expression vectors or antisense oligonucleotides in cellular models. This could facilitate the research in therapies modifying dysregulated microRNAs.
Table 3. MicroRNAs in prion-like diseases.
Table 3. MicroRNAs in prion-like diseases.
Disease Species/Model Tissue Type miRNA Main Finding References
Alzheimer’s disease HumanBrain, peripheral blood, serum, CSF, serum and CSF exosomes, and plasma extracellular vesiclesmiRNA profilesDifferential miRNA expression profiles in AD patients[111,112,113,114,115,116,117,118,119,120,121,122]
APP/PS1 and 5XFAD miceBrain and urinary exosomesmiRNA profilesDifferential miRNA expression profiles in AD mouse models[123,124,125]
HumanSerummiR-193a-3p, miR-133bNeuroprotective roles attenuating Aβ accumulation and its associated neurotoxicity[170,171]
HumanBrainmiR-335-5p, miR-361-3p[172,173]
SAMP8 miceHippocampusmiR-340[175]
HumanSH-SY5Y, SK-N-SH cellsmiR-107[176]
HumanBlood plasmamiR-200a-3p[174]
RatHippocampusmiR-134-5pInvolved in rescuing AD synaptic plasticity[177]
C57BL/6J, Tg2576 miceHippocampusmiR-124Alleviates tau pathology and mediates synaptic and memory deficits [178,180]
SAMP8 mice, HumanHippocampus, serummiR-34cMediates synaptic and memory deficits[179]
HumanBlood, BrainmiR-146a, miR-181a, miR-142-3pAssociated with AD risk and progression[181,182]
Parkinson’s disease HumanBrain, gut, plasma, serum, CSF, saliva, plasma exosomes, serum extracellular vesicles, and iPSC-derived dopaminergic neuronsmiRNA profilesDifferential miRNA expression profiles in PD patients[126,127,128,129,130,131,132,133,134,135,136,137]
[139]
RatPeripheral bloodmiRNA profileDifferential miRNA expression profile in a PD rat model[138]
HumanSerummiR-150Amelioration of PD-associated neuroinflammation[183]
C57BL/6 miceBrainmiR-let-7a, miR-190[184,185]
Human, ratSH-SY5Y, PC-12 cellsmiR-135b[186]
Wistar rats, C57BL/6 mice, HumanBrain, SH-SY5Y cellsmiR-375, miR-29cDecrease in dopaminergic neurons’ damage and neuroinflammatory response [187,188]
Sprague–Dawley ratsBrainmiR-3557, miR-324Involved in delaying PD neurodegeneration[189]
Human, ratSH-SY5Y, PC-12 cellsmiR-410Overexpression exerts neuroprotective effects against apoptosis and reactive oxygen species production[190]
C57BL/6 miceBrainmiR-326Overexpression promotes autophagy of dopaminergic neurons[191]
MouseBV2 cellsmiR-195Downregulation might induce microglia-mediated neuroinflammation activation[192]
Sprague–Dawley ratsPeripheral blood, brainmiR-7Involved in regulating brain-derived neurotrophic factor expression in early stages of PD[193]
HumanPBMCs, SH-SY5Y cellsmiR-376aImplicated in PD pathogenesis regulating the expression of mitochondrial-related genes[194]
Amyotrophic lateral sclerosis HumanBrain, spinal cord, skeletal muscle, neuromuscular junctions, plasma, serum, leukocytes, CSF, plasma, serum, brain and spinal cord extracellular vesicles, motor neuron-derived exosomes, iPSC-derived motor neurons, and motor neuron progenitorsmiRNA profilesDifferential miRNA expression profiles in ALS patients[140,141,142,143,144,145,146,147,148,149,150,151,152,153,154,155]
[158]
SOD1G86R and SOD1G93A miceSerum, skeletal musclemiRNA profilesDifferential miRNA expression profiles in ALS mouse models[156,157]
HumanSerummiR-335-5p Downregulation may enhance mitophagy, autophagy, and apoptosis pathways[195]
Wobbler miceSpinal cordmiR-375-3pRegulates target structures that intervene at the apoptosis pathway[196]
Wobbler miceCerebellummiR-29b-3pDownregulates proapoptotic factors, leading to neuroprotection[197]
SOD1G93A miceMusclemiR-126-5pInvolved in axon degeneration[198]
HumanAstrocytesmiR-494-3p[199]
HumanCNSmiR-1825[200]
SOD1 miceSpinal cordmiR-338-3pUpregulation decreases glycogenolysis causing glycogen accumulation within the spinal cord[201]
HumanSpinal cordmiR-105, miR-9Contribute to intermediate filament aggregation in ALS[202]
MouseAstrocytesmiR-218Affects astrocyte function negatively[203]
Human, SOD1G93A miceSkeletal muscle, serum, plasmamiR-206Upregulated. Involved in neuromuscular junction, regeneration, and muscle atrophy[79,156]
HumanSkeletal musclemiR-27a, miR-221, miR-155Increased in FALS patients[79]
Human, SOD1G93A miceMotor neuronsmiR-17~92 clusterAssociated with vulnerability of motor neurons to ALS-related degeneration[205]
Huntington’s disease C57BL/6, R6/1 and BACHD miceBrainmiRNA profilesDifferential miRNA expression profiles in HD mouse models[164,165,166,167]
HumanBrain, plasma, CSFmiRNA profilesDifferential miRNA expression profiles in HD patients. Association between CSF-miRNAs expression profiles and the earliest prodromal stages of HD[172,173,174,175,176]
Hdh miceStriatum, cortexmiRNA profilesmiRNA regulation may have a limited global role in responding to HD[206]
R6/2 miceBrainmiR-132Decreased levels, whose restoration confers amelioration in motor function and lifespan[207]
FVB mouse embryos, HD1/HD7/WT monkeyNeural progenitors, neural cellsmiR-196aNeuroprotective effects[208,209]
R6/2 miceNeuronal stem cellsmiR-27aReduces mutant htt aggregation[210]
Hu128/21 mice, sheepStriatumArtificial miRNAsReduce mutant htt levels[211,212]

3. Epigenetic Changes in Prion Diseases

Compared to prion-like diseases, there are very few studies on the involvement of epigenetic changes in transmissible spongiform encephalopathies.
It is known that the prion protein (PrP) is able to bind to RNA and DNA molecules [213]. As these nucleic acids can induce PrP aggregation, they have been proposed as catalysts in the conversion of the PrPC to the pathologic form PrPSc [214]. Different DNA molecules are capable of binding to recombinant PrP (rPrP), resulting in complex aggregates [215]. Interestingly, the GC content of these DNA molecules seems to be important in the binding, affinity, stability, and aggregation abilities and in the toxic species generation [215]. A recent study has evaluated the neurotoxic effect of the inoculation of a PrP-DNA complex in the lateral ventricle of Swiss mice, which, after inoculation, showed cognitive impairment, hippocampal synapse loss, and intense glial activation [216]. In contrast, in human neuroblastoma cell cultures, PrP cytotoxicity is attenuated when combined with DNA molecules, which stabilize PrP structure and reduce its pathogenic properties [217].
Different RNA molecules can also bind to PrP and trigger its aggregation and conversion to PrPSc, the efficiency of the conversion depending on the RNA source [216,218,219]. Some of these RNA molecules, specifically RNA aptamers, are able to bind and stabilize PrPC and reduce PrPSc levels in infected mouse neuronal cells [220]. Further studies are needed in order to evaluate the involvement of these nucleic acids in prion pathology and to develop potential therapeutic strategies.

3.1. DNA Methylation Profiles

DNA methylation might also be involved in the pathogenesis of prion diseases. A study of the mouse prion protein gene (PRNP) encoding the PrPC protein has reported an association between DNA methylation and PRNP gene expression. The PRNP gene promoter region seems to be unmethylated, and the methylation status of one of the PRNP enhancer regions was negatively correlated with PRNP expression [221]. In addition, during neuronal differentiation of mouse embryonic carcinoma P19C6 cells, the expression of PRNP was markedly increased, while CpG methylation was significantly reduced, suggesting that DNA methylation could be implicated in mediating PRNP expression regulation [222].
Only three studies have analyzed the methylome of prion diseases. A genome-wide methylation study has shown differentially methylated positions in blood from sporadic Creutzfeldt–Jakob disease (sCJD) patients compared with healthy controls, some of these positions correlated with disease progression [223]. Furthermore, another genome-wide methylation study of the CNS of sheep naturally infected with scrapie has identified differentially methylated regions between control and scrapie animals, belonging some of them to genes with possible neuroprotective roles and to genes that may contribute to scrapie disease progression [224]. Interestingly, a recent study was able to identify different DNA methylation patterns in tonsil and appendix lymphoreticular tissues between sCJD patients and healthy individuals [225], pointing to a potential source of diagnostic biomarkers in prion diseases.
Although these three works have been performed in different tissues (blood, CNS, and lymphoid tissues) and species (CJD human patients and scrapie-infected sheep) and have used different methodologies (450k methylation array for humans and whole genome bisulfite sequencing), we have compared the genes detected in each work containing either DMR or DMP. Twelve genes were differentially methylated in both ovine CNS and CJD blood. Further studies are necessary to confirm the role of these common genes in prion diseases. Supplementary Table S4 shows the common DMR and DMP in prion and prion-like diseases.
We have compared the methylation DMR and DMP profiles between these studies and those performed in prion-like diseases. As different methodology has been used, only DMPs obtained in CJD samples were compared with the other diseases. Only 2 DMPs identified in CJD blood were found in common with AD, 3 with PD, and 1 with ALS. However, of a total of 8907 DMRs observed in CNS of scrapie-infected sheep, 634 are also altered in PD, 60 in Alzheimer’s, 51 in ALS, and 61 in HD. This large difference is due to the fact that PD and scrapie were analyzed using the same methodology, bisulfite-treated DNA sequencing. More such studies are needed in the other prion-like diseases to determine whether or not there are common genes differentially methylated in all prion-misfolded pathologies.

3.2. Histone Modifications

Yeast ESI+, for expressed sub-telomeric information, is the prion form of the Set3C histone deacetylase scaffold Snt1 (NCOR1 in humans). This prion, in response to cell cycle arrest, is able to activate gene expression through H4 acetylation and RNA polymerase II recruitment [226]. Other histone deacetylases, namely HDAC6 and sirtuin-1 (SIRT1), have shown protective effects during prion infection. Although many studies in prion-like diseases are addressed to inhibit HDAC6 as a potential therapeutic approach, in cerebral cortical neurons, overexpression of HDAC6 alleviates prion peptide-mediated neuronal cell death and toxicity [227]. On the other hand, overexpression of SIRT1, which is decreased in the brains of scrapie-infected rodents and in prion-infected SMB-S15 cells, reduces PrPSc levels and protects against prion protein-induced neuronal cell death and mitochondrial dysfunction [228,229,230]. Therefore, although HDAC inhibition is proposed as a potential therapeutic strategy in prion-like diseases, this approach does not seem the best choice for treating prion diseases. Further analysis will be necessary to find out the role of other HDACs in prion diseases and to design novel therapies that maybe could be focused on the overexpression of HDACs.

3.3. MicroRNA Profiles

A number of studies have reported changes in miRNA expression profiles during prion infection in the CNS, plasma, and synaptoneurosomes of preclinical and clinical scrapie-infected mice [231,232,233], in the serum of elk infected with the chronic wasting disease (CWD) [234] and in plasma of naturally infected classical scrapie sheep [235]. When comparing these profiles with the ones reported in prion-like diseases, there are some miRNAs commonly altered in the two groups of diseases. Table 4 shows the common upregulated, and downregulated miRNAs, and Supplementary Table S3 lists the datasets generated from the different miRNA profile studies in prion diseases. The prion-like disease with more miRNAs in common with prion diseases is ALS, whereas the one with fewer miRNAs in common is HD. More studies are required in order to find specific miRNAs that are only altered in prion diseases, even only in each type of prion disease, specifically for their use as diagnostic biomarkers.
Although the potential role of most of these alterations in TSE pathology is unknown, multiple miRNAs regulate PrPC levels both directly and indirectly in human neuroectodermal cell lines [236]. On the other hand, miR-16, which is increased in hippocampal neurons during presymptomatic prion disease, could decrease neurite length and branching, probably via the downregulation of components of the MAPK/ERK pathway [237]. Additionally, a single nucleotide polymorphism in miR-146a has been associated with susceptibility to FFI (fatal familial insomnia) and with the appearance of some clinical features in sCJD patients [238]. Interestingly, artificial miRNAs have also been used to reduce PrPC and subsequently suppress PrPSc propagation in primary mixed neuronal and glial cells culture [239].

4. Concluding Remarks

It is evident that epigenetic mechanisms, namely DNA methylation, histone post-translational modifications, and microRNAs, are involved in the pathophysiology of neurodegenerative diseases. Increasing evidence in prion-like diseases and, more recently, in prion diseases has shown that epigenetic modifications can modulate different pathogenic mechanisms occurring in these neurodegenerative disorders. Nevertheless, these epigenetic mechanisms seem to act differently in each of these diseases. Global DNA methylation changes have been detected in prion diseases and prion-like diseases in a variety of tissues and in several specific genes, showing different trends in the global methylation profile of each disease and several genes harboring differentially methylated regions and positions that match between some of these diseases. Modulation of HDAC enzymes seems to also be a common epigenetic mechanism in these diseases. Interestingly, among all the HDACs, the enzyme HDAC6 is involved in both prion diseases and prion-like diseases, although the mechanism of action is different between the two groups of diseases. On the other hand, other HDCAs, namely HDAC2, HDAC3, and HDAC4, are associated with different aspects of prion-like diseases. Additionally, miRNA profiles seem to be the most specific of each disease. However, there are two miRNAs, miR-144-3p and miR-22-5p, that seem to be commonly upregulated in the four prion-like diseases but not in prion diseases, and other miRNAs, such as miR-335-5p, miR-375, and miR-27a, have functions in AD, PD, HD, and ALS, but they seem to regulate different pathways in each disease. Prion and prion-like diseases also share several miRNAs in common, ALS being the prion-like disease with the highest number of common miRNAs. In addition, artificial miRNAs have successfully been used in both HD and prion diseases to reduce misfolded protein levels. Further research is still needed, especially in prion diseases where knowledge is still scarce, in order to elucidate the exact molecular pathways by which these epigenetic mechanisms perform their regulatory roles and to identify potential epigenetic diagnostic and therapeutic biomarkers.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ijms232012609/s1. References [19,20,21,22,23,24,25,26,27,28,29,30,31,32,33,34,35,36,37,111,112,113,114,115,116,117,118,119,120,121,122,126,127,128,129,130,131,132,133,134,135,136,137,140,141,142,143,144,145,146,147,148,149,150,151,152,153,154,155,159,160,161,162,163,231,232,233,234,235] are cited in the Supplementary Materials.

Author Contributions

Conceptualization, A.H. and I.M.-B.; writing—original draft preparation, A.H.; writing—review and editing, I.M.-B. and J.M.T.; visualization, A.H.; supervision, I.M.-B., J.M.T., and R.B.; funding acquisition, I.M.-B. and R.B. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by Gobierno de Aragón and the European Social Fund co-financed predoctoral grant Order IIU/2023/2017, AGL2015-67945-P funded by MINECO, reference group A19-20R funded by Gobierno de Aragón co-financed with FEDER 2014-2020 Construyendo Europa desde Aragón and the European Regional Development Fund (ERDF).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Moore, L.D.; Le, T.; Fan, G. DNA Methylation and Its Basic Function. Neuropsychopharmacology 2013, 38, 23–38. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Audia, J.E.; Campbell, R.M. Histone Modifications and Cancer. Cold Spring Harb. Perspect. Biol. 2016, 8, a019521. [Google Scholar] [CrossRef] [PubMed]
  3. Zhang, J.; Jing, L.; Li, M.; He, L.; Guo, Z. Regulation of histone arginine methylation/demethylation by methylase and demethylase (Review). Mol. Med. Rep. 2019, 19, 3963–3971. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Huang, W. MicroRNAs: Biomarkers, diagnostics, and therapeutics. Methods Mol. Biol. 2017, 1617, 57–67. [Google Scholar]
  5. Place, R.F.; Li, L.C.; Pookot, D.; Noonan, E.J.; Dahiya, R. MicroRNA-373 induces expression of genes with complementary promoter sequences. Proc. Natl. Acad. Sci. USA 2008, 105, 1608–1613. [Google Scholar] [CrossRef] [Green Version]
  6. Bottani, M.; Banfi, G.; Lombardi, G. Perspectives on miRNAs as Epigenetic Markers in Osteoporosis and Bone Fracture Risk: A Step Forward in Personalized Diagnosis. Front. Genet. 2019, 10, 1044. [Google Scholar] [CrossRef]
  7. Lardenoije, R.; Iatrou, A.; Kenis, G.; Kompotis, K.; Steinbusch, H.W.; Mastroeni, D.; Coleman, P.; Lemere, C.A.; Hof, P.R.; van den Hove, D.L.A.; et al. The epigenetics of aging and neurodegeneration. Prog. Neurobiol. 2015, 131, 21–64. [Google Scholar] [CrossRef]
  8. Hwang, J.Y.; Aromolaran, K.A.; Zukin, R.S. The emerging field of epigenetics in neurodegeneration and neuroprotection. Nat. Rev. Neurosci. 2017, 18, 347–361. [Google Scholar] [CrossRef]
  9. Heyward, F.D.; Sweatt, J.D. DNA Methylation in Memory Formation: Emerging Insights. Neuroscientist 2015, 21, 475–489. [Google Scholar] [CrossRef] [Green Version]
  10. Miller, C.A.; Sweatt, J.D. Covalent Modification of DNA Regulates Memory Formation. Neuron 2007, 53, 857–869. [Google Scholar] [CrossRef] [Green Version]
  11. Hu, Z.; Li, Z. miRNAs in synapse development and synaptic plasticity. Curr. Opin. Neurobiol. 2017, 45, 24–31. [Google Scholar] [CrossRef] [PubMed]
  12. Woldemichael, B.T.; Mansuy, I.M. Micro-RNAs in cognition and cognitive disorders: Potential for novel biomarkers and therapeutics. Biochem. Pharmacol. 2016, 104, 1–7. [Google Scholar] [CrossRef] [PubMed]
  13. Aksoy-Aksel, A.; Zampa, F.; Schratt, G. MicroRNAs and synaptic plasticity-a mutual relationship. Philos. Trans. R. Soc. B Biol. Sci. 2014, 369, 20130515. [Google Scholar] [CrossRef] [Green Version]
  14. Jaunmuktane, Z.; Brandner, S. Invited Review: The role of prion-like mechanisms in neurodegenerative diseases. Neuropathol. Appl. Neurobiol. 2019, 46, 522–545. [Google Scholar] [CrossRef] [Green Version]
  15. Masnata, M.; Sciacca, G.; Maxan, A.; Bousset, L.; Denis, H.L.; Lauruol, F.; David, L.; Saint-Pierre, M.; Kordower, J.H.; Melki, R.; et al. Demonstration of prion-like properties of mutant huntingtin fibrils in both in vitro and in vivo paradigms. Acta Neuropathol. 2019, 137, 981–1001. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Ma, J.; Wang, F. Prion disease and the ‘protein-only hypothesis’. Essays Biochem. 2014, 56, 181–191. [Google Scholar] [CrossRef]
  17. Prusiner, S. Novel proteinaceous infectious particles cause scrapie. Science 1982, 216, 136–144. [Google Scholar] [CrossRef] [Green Version]
  18. Chakravarty, A.K.; Jarosz, D.F. More than Just a Phase: Prions at the Crossroads of Epigenetic Inheritance and Evolutionary Change. J. Mol. Biol. 2018, 430, 4607–4618. [Google Scholar] [CrossRef]
  19. Li, H.; Guo, Z.; Guo, Y.; Li, M.; Yan, H.; Cheng, J.; Wang, C.; Hong, G. Common DNA methylation alterations of Alzheimer’s disease and aging in peripheral whole blood. Oncotarget 2016, 7, 19089–19098. [Google Scholar] [CrossRef] [Green Version]
  20. Li, Q.S.; Vasanthakumar, A.; Davis, J.W.; Idler, K.B.; Nho, K.; Waring, J.F.; Saykin, A.J.; for the Alzheimer’s Disease Neuroimaging Initiative (ADNI). Association of peripheral blood DNA methylation level with Alzheimer’s disease progression. Clin. Epigenetics 2021, 13, 191. [Google Scholar] [CrossRef]
  21. Wang, C.; Chen, L.; Yang, Y.; Zhang, M.; Wong, G. Identification of potential blood biomarkers for Parkinson’s disease by gene expression and DNA methylation data integration analysis. Clin. Epigenet. 2019, 11, 24. [Google Scholar] [CrossRef] [PubMed]
  22. Henderson, A.R.; Wang, Q.; Meechoovet, B.; Siniard, A.L.; Naymik, M.; de Both, M.; Huentelman, M.J.; Caselli, R.J.; Driver-Dunckley, E.; Dunckley, T. DNA Methylation and Expression Profiles of Whole Blood in Parkinson’s Disease. Front. Genet. 2021, 12, 640266. [Google Scholar] [CrossRef] [PubMed]
  23. Chuang, Y.-H.; Paul, K.C.; Bronstein, J.M.; Bordelon, Y.; Horvath, S.; Ritz, B. Parkinson’s disease is associated with DNA methylation levels in human blood and saliva. Genome Med. 2017, 9, 76. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Hop, P.J.; Zwamborn, R.A.; Hannon, E.; Shireby, G.L.; Nabais, M.F.; Walker, E.M.; van Rheenen, W.; van Vugt, J.J.; Dekker, A.M.; Westeneng, H.-J.; et al. Genome-wide study of DNA methylation shows alterations in metabolic, inflammatory, and cholesterol pathways in ALS. Sci. Transl. Med. 2022, 14, eabj0264. [Google Scholar] [CrossRef]
  25. Zadel, M.; Maver, A.; Kovanda, A.; Peterlin, B. DNA methylation profiles in whole blood of Huntington’s disease patients. Front. Neurol. 2018, 9, 655. [Google Scholar] [CrossRef] [Green Version]
  26. Lu, A.T.; Narayan, P.; Grant, M.J.; Langfelder, P.; Wang, N.; Kwak, S.; Wilkinson, H.; Chen, R.Z.; Chen, J.; Bawden, C.S.; et al. DNA methylation study of Huntington’s disease and motor progression in patients and in animal models. Nat. Commun. 2020, 11, 4529. [Google Scholar] [CrossRef]
  27. Zhao, J.; Zhu, Y.; Yang, J.; Li, L.; Wu, H.; de Jager, P.L.; Jin, P.; Bennett, D.A. A genome-wide profiling of brain DNA hydroxymethylation in Alzheimer’s disease. Alzheimer’s Dement. 2017, 13, 674–688. [Google Scholar] [CrossRef]
  28. Watson, C.T.; Roussos, P.; Garg, P.; Ho, D.J.; Azam, N.; Katsel, P.L.; Haroutunian, V.; Sharp, A.J. Genome-wide12 DNA methylation profiling in the superior temporal gyrus reveals epigenetic signatures associated with Alzheimer’s disease. Genome Med. 2016, 8, 5. [Google Scholar] [CrossRef] [Green Version]
  29. Gao, Z.; Fu, H.; Zhao, L.; Sun, Z.; Yang, Y.; Zhu, H. Aberrant DNA methylation associated with Alzheimer’s disease in the superior temporal gyrus. Exp. Ther. Med. 2017, 15, 103–108. [Google Scholar] [CrossRef]
  30. Altuna, M.; Casado, A.U.; de Gordoa, J.S.-R.; Zelaya, M.V.; Labarga, A.; Lepesant, J.M.J.; Roldán, M.; Blanco-Luquin, I.; Perdones, Á.; Larumbe, R.; et al. DNA methylation signature of human hippocampus in Alzheimer’s disease is linked to neurogenesis. Clin. Epigenetics 2019, 11, 91. [Google Scholar] [CrossRef] [Green Version]
  31. Smith, A.; Smith, R.G.; Pishva, E.; Hannon, E.; Roubroeks, J.A.Y.; Burrage, J.; Troakes, C.; Al-Sarraj, S.; Sloan, C.; Mill, J.; et al. Parallel profiling of DNA methylation and hydroxymethylation highlights neuropathology-associated epigenetic variation in Alzheimer’s disease. Clin. Epigenetics 2019, 11, 52. [Google Scholar] [CrossRef] [PubMed]
  32. Young, J.I.; Sivasankaran, S.K.; Wang, L.; Ali, A.; Mehta, A.; Davis, D.A.; Dykxhoorn, D.M.; Petito, C.K.; Beecham, G.W.; Martin, E.R.; et al. Genome-wide brain DNA methylation analysis suggests epigenetic reprogramming in Parkinson disease. Neurol. Genet. 2019, 5, e342. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Appleby-Mallinder, C.; Schaber, E.; Kirby, J.; Shaw, P.; Cooper-Knock, J.; Heath, P.R.; Highley, J.R. TDP43 proteinopathy is associated with aberrant DNA methylation in human amyotrophic lateral sclerosis. Neuropathol. Appl. Neurobiol. 2020, 47, 61–72. [Google Scholar] [CrossRef] [PubMed]
  34. Wang, F.; Yang, Y.; Lin, X.; Wang, J.-Q.; Wu, Y.-S.; Xie, W.; Wang, D.; Zhu, S.; Liao, Y.-Q.; Sun, Q.; et al. Genome-wide loss of 5-hmC is a novel epigenetic feature of Huntington’s disease. Hum. Mol. Genet. 2013, 22, 3641–3653. [Google Scholar] [CrossRef]
  35. Horvath, S.; Langfelder, P.; Kwak, S.; Aaronson, J.; Rosinski, J.; Vogt, T.F.; Eszes, M.; Faull, R.L.M.; Curtis, M.A.; Waldvogel, H.J.; et al. Huntington’s disease accelerates epigenetic aging of human brain and disrupts DNA methylation levels. Aging 2016, 8, 1485–1512. [Google Scholar] [CrossRef] [Green Version]
  36. Fernández-Santiago, R.; Carballo-Carbajal, I.; Castellano, G.; Torrent, R.; Richaud, Y.; Sánchez-Danés, A.; Vilarrasa-Blasi, R.; Sánchez-Pla, A.; Mosquera, J.L.; Soriano, J.; et al. Aberrant epigenome in iPSC-derived dopaminergic neurons from Parkinson’s disease patients. EMBO Mol. Med. 2015, 7, 1529–1546. [Google Scholar] [CrossRef]
  37. Fernández-Santiago, R.; Merkel, A.; Castellano, G.; Heath, S.; Raya, A.; Tolosa, E.; Martí, M.J.; Consiglio, A.; Ezquerra, M. Whole-genome DNA hyper-methylation in iPSC-derived dopaminergic neurons from Parkinson’s disease patients. Clin. Epigenet. 2019, 11, 108. [Google Scholar] [CrossRef] [Green Version]
  38. Heberle, H.; Meirelles, G.V.; Da Silva, F.R.; Telles, G.P.; Minghim, R. InteractiVenn: A Web-Based Tool for the Analysis of Sets through Venn Diagrams. BMC Bioinform. 2015, 16, 169. [Google Scholar] [CrossRef] [Green Version]
  39. De Souza, R.A.; Islam, S.A.; McEwen, L.M.; Mathelier, A.; Hill, A.; Mah, S.M.; Wasserman, W.W.; Kobor, M.S.; Leavitt, B.R. DNA methylation profiling in human Huntington’s disease brain. Hum. Mol. Genet. 2016, 25, 2013–2030. [Google Scholar] [CrossRef] [Green Version]
  40. Horvath, S. DNA methylation age of human tissues and cell types. Genome Biol. 2013, 14, R115. [Google Scholar]
  41. Li, P.; Marshall, L.; Oh, G.; Jakubowski, J.L.; Groot, D.; He, Y.; Wang, T.; Petronis, A.; Labrie, V. Epigenetic dysregulation of enhancers in neurons is associated with Alzheimer’s disease pathology and cognitive symptoms. Nat. Commun. 2019, 10, 2246. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Sanchez-Mut, J.V.; Glauser, L.; Monk, D.; Gräff, J. Comprehensive analysis of PM20D1 QTL in Alzheimer’s disease. Clin. Epigenet. 2020, 12, 20. [Google Scholar] [CrossRef] [PubMed]
  43. Eryilmaz, I.E.; Cecener, G.; Erer, S.; Egeli, U.; Tunca, B.; Zarifoglu, M.; Elibol, B.; Tokcaer, A.B.; Saka, E.; Demirkiran, M.; et al. Epigenetic approach to early-onset Parkinson’s disease: Low methylation status of SNCA and PARK2 promoter regions. Neurol. Res. 2017, 39, 965–972. [Google Scholar] [CrossRef]
  44. Taskesen, E.; Mishra, A.; van der Sluis, S.; Ferrari, R.; International FTD-Genomics Consortium; Veldink, J.H.; van Es, M.A.; Smit, A.B.; Posthuma, D.; Pijnenburg, Y. Susceptible genes and disease mechanisms identified in frontotemporal dementia and frontotemporal dementia with Amyotrophic Lateral Sclerosis by DNA-methylation and GWAS. Sci. Rep. 2017, 7, 8899. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Ebbert, M.; Ross, C.A.; Pregent, L.J.; Lank, R.J.; Zhang, C.; Katzman, R.B.; Jansen-West, K.; Song, Y.; da Rocha, E.L.; Palmucci, C.; et al. Conserved DNA methylation combined with differential frontal cortex and cerebellar expression distinguishes C9orf72-associated and sporadic ALS, and implicates SERPINA1 in disease. Acta Neuropathol. 2017, 134, 715–728. [Google Scholar] [CrossRef]
  46. Kim, B.W.; Jeong, Y.E.; Wong, M.; Martin, L.J. DNA damage accumulates and responses are engaged in human ALS brain and spinal motor neurons and DNA repair is activatable in iPSC-derived motor neurons with SOD1 mutations. Acta Neuropathol. Commun. 2020, 8, 7. [Google Scholar] [CrossRef] [Green Version]
  47. Bai, G.; Cheung, I.; Shulha, H.P.; Coelho, J.E.; Li, P.; Dong, X.; Jakovcevski, M.; Wang, Y.; Grigorenko, A.; Jiang, Y.; et al. Epigenetic dysregulation of hairy and enhancer of split 4 (HES4) is associated with striatal degeneration in postmortem Huntington brains. Hum. Mol. Genet. 2014, 24, 1441–1456. [Google Scholar] [CrossRef] [Green Version]
  48. Gutierrez, A.; Corey-Bloom, J.; Thomas, E.A.; Desplats, P. Evaluation of Biochemical and Epigenetic Measures of Peripheral Brain-Derived Neurotrophic Factor (BDNF) as a Biomarker in Huntington’s Disease Patients. Front. Mol. Neurosci. 2020, 12, 335. [Google Scholar] [CrossRef] [Green Version]
  49. Bachman, M.; Uribe-Lewis, S.; Yang, X.; Williams, M.; Murrell, A.; Balasubramanian, S. 5-Hydroxymethylcytosine is a predominantly stable DNA modification. Nat. Chem. 2014, 6, 1049–1055. [Google Scholar] [CrossRef] [Green Version]
  50. Ellison, E.M.; Abner, E.L.; Lovell, M.A. Multiregional analysis of global 5-methylcytosine and 5-hydroxymethylcytosine throughout the progression of Alzheimer’s disease. J. Neurochem. 2016, 140, 383–394. [Google Scholar] [CrossRef] [Green Version]
  51. Kaut, O.; Kuchelmeister, K.; Moehl, C.; Wüllner, U. 5-methylcytosine and 5-hydroxymethylcytosine in brains of patients with multiple system atrophy and patients with Parkinson’s disease. J. Chem. Neuroanat. 2019, 96, 41–48. [Google Scholar] [CrossRef] [PubMed]
  52. Laszlo, A.H.; Derrington, I.M.; Brinkerhoff, H.; Langford, K.W.; Nova, I.C.; Samson, J.M.; Bartlett, J.J.; Pavlenok, M.; Gundlach, J.H. Detection and mapping of 5-methylcytosine and 5-hydroxymethylcytosine with nanopore MspA. Proc. Natl. Acad. Sci. USA 2013, 110, 18904–18909. [Google Scholar] [CrossRef] [PubMed]
  53. Hamzeiy, H.; Savaş, D.; Tunca, C.; Şen, N.E.; Gündoǧdu Eken, A.; Şahbaz, I.; Calini, D.; Tiloca, C.; Ticozzi, N.; Ratti, A.; et al. Elevated Global DNA Methylation Is Not Exclusive to Amyotrophic Lateral Sclerosis and Is Also Ob-served in Spinocerebellar Ataxia Types 1 and 2. Neurodegener. Dis. 2018, 18, 38–48. [Google Scholar] [CrossRef] [PubMed]
  54. Jackson, J.L.; Finch, N.A.; Baker, M.C.; Kachergus, J.M.; DeJesus-Hernandez, M.; Pereira, K.; Christopher, E.; Prudencio, M.; Heckman, M.G.; Thompson, E.A.; et al. Elevated methylation levels, reduced expression levels, and frequent contractions in a clinical cohort of C9orf72 expansion carriers. Mol. Neurodegener. 2020, 15, 7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Zhang, M.; Tartaglia, M.C.; Moreno, D.; Sato, C.; McKeever, P.; Weichert, A.; Keith, J.; Robertson, J.; Zinman, L.; Rogaeva, E. DNA methylation age-acceleration is associated with disease duration and age at onset in C9orf72 patients. Acta. Neuropathol. 2017, 134, 271–279. [Google Scholar] [CrossRef] [Green Version]
  56. Coppedè, F.; Stoccoro, A.; Mosca, L.; Gallo, R.; Tarlarini, C.; Lunetta, C.; Marocchi, A.; Migliore, L.; Penco, S. Increase in DNA methylation in patients with amyotrophic lateral sclerosis carriers of not fully penetrant SOD1 mutations. Amyotroph. Lateral Scler. Front. Degener. 2018, 19, 93–101. [Google Scholar] [CrossRef]
  57. Madrid, A.; Hogan, K.J.; Papale, L.A.; Clark, L.R.; Asthana, S.; Johnson, S.C.; Alisch, R.S. DNA Hypomethylation in Blood Links B3GALT4 and ZADH2 to Alzheimer’s Disease. J. Alzheimer’s Dis. 2018, 66, 927–934. [Google Scholar] [CrossRef]
  58. Salcedo-Tacuma, D.; Melgarejo, J.D.; Mahecha, M.F.; Ortega-Rojas, J.; Arboleda-Bustos, C.E.; Pardo-Turriago, R.; Arboleda, H. Differential Methylation Levels in CpGs of the BIN1 Gene in Individuals with Alzheimer Disease. Alzheimer Dis. Assoc. Disord. 2019, 33, 321–326. [Google Scholar] [CrossRef]
  59. Li, K.-X.; Sun, Q.; Wei, L.-L.; Du, G.-H.; Huang, X.; Wang, J.-K. ERα Gene Promoter Methylation in Cognitive Function and Quality of Life of Patients with Alzheimer Disease. J. Geriatr. Psychiatry Neurol. 2019, 32, 221–228. [Google Scholar] [CrossRef]
  60. Mao, W.; Zhao, C.; Ding, H.; Liang, K.; Xue, J.; Chan, P.; Cai, Y. Pyrosequencing analysis of methylation levels of clock genes in leukocytes from Parkinson’s disease patients. Neurosci. Lett. 2018, 668, 115–119. [Google Scholar] [CrossRef]
  61. Shao, Y.; Shaw, M.; Todd, K.; Khrestian, M.; D’Aleo, G.; Barnard, P.J.; Zahratka, J.; Pillai, J.; Yu, C.-E.; Keene, C.D.; et al. DNA methylation of TOMM40-APOE-APOC2 in Alzheimer’s disease. J. Hum. Genet. 2018, 63, 459–471. [Google Scholar] [CrossRef] [PubMed]
  62. Kobayashi, N.; Shinagawa, S.; Nagata, T.; Shimada, K.; Shibata, N.; Ohnuma, T.; Kasanuki, K.; Arai, H.; Yamada, H.; Nakayama, K.; et al. Usefulness of DNA Methylation Levels in COASY and SPINT1 Gene Promoter Regions as Biomarkers in Diagnosis of Alzheimer’s Disease and Amnestic Mild Cognitive Impairment. PLoS ONE 2016, 11, e0168816. [Google Scholar] [CrossRef] [PubMed]
  63. Ozaki, Y.; Yoshino, Y.; Yamazaki, K.; Ochi, S.; Iga, J.; Nagai, M.; Nomoto, M.; Ueno, S. DRD2 methylation to differentiate dementia with Lewy bodies from Parkinson’s disease. Acta Neurol. Scand. 2019, 141, 177–182. [Google Scholar] [CrossRef] [PubMed]
  64. Cohen-Hadad, Y.; Altarescu, G.; Eldar-Geva, T.; Levi-Lahad, E.; Zhang, M.; Rogaeva, E.; Gotkine, M.; Bartok, O.; Ashwal-Fluss, R.; Kadener, S.; et al. Marked Differences in C9orf72 Methylation Status and Isoform Expression between C9/ALS Human Embryonic and Induced Pluripotent Stem Cells. Stem Cell Rep. 2016, 7, 927–940. [Google Scholar] [CrossRef] [Green Version]
  65. Kantor, B.; Tagliafierro, L.; Gu, J.; Zamora, M.E.; Ilich, E.; Grenier, C.; Huang, Z.Y.; Murphy, S.; Chiba-Falek, O. Downregulation of SNCA Expression by Targeted Editing of DNA Methylation: A Potential Strategy for Precision Therapy in PD. Mol. Ther. 2018, 26, 2638–2649. [Google Scholar] [CrossRef] [Green Version]
  66. Park, H.; Shin, J.; Kim, Y.; Saito, T.; Saido, T.C.; Kim, J. CRISPR/dCas9-Dnmt3a-mediated targeted DNA methylation of APP rescues brain pathology in a mouse model of Alzheimer’s disease. Transl. Neurodegener. 2022, 11, 41. [Google Scholar] [CrossRef]
  67. Stoccoro, A.; Mosca, L.; Carnicelli, V.; Cavallari, U.; Lunetta, C.; Marocchi, A.; Migliore, L.; Coppedè, F. Mitochondrial DNA copy number and D-loop region methylation in carriers of amyotrophic lateral sclerosis gene mutations. Epigenomics 2018, 10, 1431–1443. [Google Scholar] [CrossRef]
  68. Xu, Y.; Xu, L.; Han, M.; Liu, X.; Li, F.; Zhou, X.; Wang, Y.; Bi, J. Altered mitochondrial DNA methylation and mitochondrial DNA copy number in an APP/PS1 transgenic mouse model of Alzheimer disease. Biochem. Biophys. Res. Commun. 2019, 520, 41–46. [Google Scholar] [CrossRef]
  69. Marzi, S.; Leung, S.K.; Ribarska, T.; Hannon, E.; Smith, A.R.; Pishva, E.; Poschmann, J.; Moore, K.; Troakes, C.; Al-Sarraj, S.; et al. A histone acetylome-wide association study of Alzheimer’s disease identifies disease-associated H3K27ac differences in the entorhinal cortex. Nat. Neurosci. 2018, 21, 1618–1627. [Google Scholar] [CrossRef] [Green Version]
  70. Klein, H.-U.; McCabe, C.; Gjoneska, E.; Sullivan, S.E.; Kaskow, B.J.; Tang, A.; Smith, R.V.; Xu, J.; Pfenning, A.R.; Bernstein, B.E.; et al. Epigenome-wide study uncovers large-scale changes in histone acetylation driven by tau pathology in aging and Alzheimer’s human brains. Nat. Neurosci. 2018, 22, 37–46. [Google Scholar] [CrossRef]
  71. Park, G.; Tan, J.; Garcia, G.; Kang, Y.; Salvesen, G.; Zhang, Z. Regulation of Histone Acetylation by Autophagy in Parkinson Disease. J. Biol. Chem. 2016, 291, 3531–3540. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Yakhine-Diop, S.M.S.; Niso-Santano, M.; Rodríguez-Arribas, M.; Gómez-Sánchez, R.; Martínez-Chacón, G.; Uribe-Carretero, E.; Navarro-García, J.A.; Ruiz-Hurtado, G.; Aiastui, A.; Cooper, J.M.; et al. Impaired Mitophagy and Protein Acetylation Levels in Fibroblasts from Parkinson’s Disease Patients. Mol. Neurobiol. 2019, 56, 2466–2481. [Google Scholar] [CrossRef] [PubMed]
  73. Schueller, E.; Paiva, I.; Blanc, F.; Wang, X.L.; Cassel, J.C.; Boutillier, A.L.; Bousiges, O. Dysregulation of histone acetylation pathways in hippocampus and frontal cortex of Alzheimer’s disease patients. Eur. Neuropsychopharmacol. 2020, 33, 101–116. [Google Scholar] [CrossRef] [PubMed]
  74. Mahady, L.; Nadeem, M.; Malek-Ahmadi, M.; Chen, K.; Perez, S.E.; Mufson, E.J. HDAC 2 dysregulation in the nucleus basalis of Meynert during the progression of Alzheimer’s disease. Neuropathol. Appl. Neurobiol. 2019, 45, 380–397. [Google Scholar] [CrossRef] [PubMed]
  75. Tan, Y.; Delvaux, E.; Nolz, J.; Coleman, P.D.; Chen, S.; Mastroeni, D. Upregulation of histone deacetylase 2 in laser capture nigral microglia in Parkinson’s disease. Neurobiol. Aging 2018, 68, 134–141. [Google Scholar] [CrossRef]
  76. Wu, Q.; Yang, X.; Zhang, L.; Zhang, Y.; Feng, L. Nuclear Accumulation of Histone Deacetylase 4 (HDAC4) Exerts Neurotoxicity in Models of Parkinson’s Disease. Mol. Neurobiol. 2017, 54, 6970–6983. [Google Scholar] [CrossRef]
  77. Buonvicino, D.; Felici, R.; Ranieri, G.; Caramelli, R.; Lapucci, A.; Cavone, L.; Muzzi, M.; Di Pietro, L.; Bernardini, C.; Zwergel, C.; et al. Effects of Class II-Selective Histone Deacetylase Inhibitor on Neuromuscular Function and Disease Progression in SOD1-ALS Mice. Neuroscience 2018, 379, 228–238. [Google Scholar] [CrossRef]
  78. Lapucci, A.; Cavone, L.; Buonvicino, D.; Felici, R.; Gerace, E.; Zwergel, C.; Valente, S.; Mai, A.; Chiarugi, A. Effect of Class II HDAC inhibition on glutamate transporter expression and survival in SOD1-ALS mice. Neurosci. Lett. 2017, 656, 120–125. [Google Scholar] [CrossRef]
  79. Pegoraro, V.; Marozzo, R.; Angelini, C. MicroRNAs and HDAC4 protein expression in the skeletal muscle of ALS patients. Clin. Neuropathol. 2020, 39, 105–114. [Google Scholar] [CrossRef]
  80. Pigna, E.; Simonazzi, E.; Sanna, K.; Bernadzki, K.M.; Proszynski, T.; Heil, C.; Palacios, D.; Adamo, S.; Moresi, V. Histone deacetylase 4 protects from denervation and skeletal muscle atrophy in a murine model of amyo-trophic lateral sclerosis. eBioMedicine 2019, 40, 717–732. [Google Scholar] [CrossRef] [Green Version]
  81. Dios, A.M.; Babu, S.; Granucci, E.J.; Mueller, K.; Mills, A.; Alshikho, M.J.; Zürcher, N.R.; Cernasov, P.; Gilbert, T.; Glass, J.D.; et al. Class I and II histone deacetylase expression is not altered in human amyotrophic lateral sclerosis: Neuropathological and positron emission tomography molecular neuroimaging evidence. Muscle Nerve 2019, 60, 443–452. [Google Scholar] [CrossRef] [PubMed]
  82. Boutillier, A.-L.; Tzeplaeff, L.; Dupuis, L. The dark side of HDAC inhibition in ALS. eBioMedicine 2019, 41, 38–39. [Google Scholar] [CrossRef]
  83. Janczura, K.J.; Volmar, C.H.; Sartor, G.C.; Rao, S.J.; Ricciardi, N.R.; Lambert, G.; Brothers, S.P.; Wahlestedt, C. Inhibition of HDAC3 reverses Alzheimer’s disease-related pathologies in vitro and in the 3xTg-AD mouse model. Proc. Natl. Acad. Sci. USA 2018, 115, E11148–E11157. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Esteves, A.; Palma, A.; Gomes, R.; Santos, D.; Silva, D.; Cardoso, S. Acetylation as a major determinant to microtubule-dependent autophagy: Relevance to Alzheimer’s and Parkinson disease pathology. Biochim. Biophys. Acta (BBA) Mol. Basis Dis. 2018, 1865, 2008–2023. [Google Scholar] [CrossRef] [PubMed]
  85. Choi, H.; Kim, H.J.; Yang, J.; Chae, S.; Lee, W.; Chung, S.; Kim, J.; Choi, H.; Song, H.; Lee, C.K.; et al. Acetylation changes tau interactome to degrade tau in Alzheimer’s disease animal and organoid models. Aging Cell. 2020, 19, e13081. [Google Scholar] [CrossRef]
  86. Jian, W.; Wei, X.; Chen, L.; Wang, Z.; Sun, Y.; Zhu, S.; Lou, H.; Yan, S.; Li, X.; Zhou, J.; et al. Inhibition of HDAC6 increases acetylation of peroxiredoxin1/2 and ameliorates 6-OHDA induced dopaminergic injury. Neurosci. Lett. 2017, 658, 114–120. [Google Scholar] [CrossRef]
  87. Kim, T.; Song, S.; Park, Y.; Kang, S.; Seo, H. HDAC inhibition by valproic acid induces neuroprotection and improvement of PD-like behaviors in LRRK2 R1441G transgenic mice. Exp. Neurobiol. 2019, 28, 504–515. [Google Scholar] [CrossRef] [Green Version]
  88. Harrison, I.F.; Smith, A.D.; Dexter, D.T. Pathological histone acetylation in Parkinson’s disease: Neuroprotection and inhibition of microglial activation through SIRT 2 inhibition. Neurosci. Lett. 2017, 666, 48–57. [Google Scholar] [CrossRef]
  89. Esteves, A.R.; Arduíno, D.M.; Silva, D.F.; Viana, S.D.; Pereira, F.C.; Cardoso, S.M. Mitochondrial Metabolism Regulates Microtubule Acetylome and Autophagy Trough Sirtuin-2: Impact for Parkinson’s Disease. Mol. Neurobiol. 2018, 55, 1440–1462. [Google Scholar] [CrossRef]
  90. Kuta, R.; Larochelle, N.; Fernandez, M.; Pal, A.; Minotti, S.; Tibshirani, M.; St.Louis, K.; Gentil, B.J.; Nalbantoglu, J.N.; Hermann, A.; et al. Depending on the stress, histone deacetylase inhibitors act as heat shock protein co-inducers in motor neurons and potentiate arimoclomol, exerting neuroprotection through multiple mechanisms in ALS models. Cell Stress Chaperones 2020, 25, 173–191. [Google Scholar] [CrossRef] [Green Version]
  91. Rossaert, E.; Pollari, E.; Jaspers, T.; van Helleputte, L.; Jarpe, M.; van Damme, P.; de Bock, K.; Moisse, M.; van den Bosch, L. Restoration of histone acetylation ameliorates disease and metabolic abnormalities in a FUS mouse model. Acta Neuropathol. Commun. 2019, 7, 107. [Google Scholar] [CrossRef]
  92. Guo, W.; Naujock, M.; Fumagalli, L.; Vandoorne, T.; Baatsen, P.; Boon, R.; Ordovás, L.; Patel, A.; Welters, M.; Vanwelden, T.; et al. HDAC6 inhibition reverses axonal transport defects in motor neurons derived from FUS-ALS patients. Nat. Commun. 2017, 8, 861. [Google Scholar] [CrossRef] [PubMed]
  93. Guo, W.; Bosch, L.V.D. Therapeutic potential of HDAC6 in amyotrophic lateral sclerosis. Cell Stress 2018, 2, 14–16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Siebzehnrübl, F.A.; Raber, K.A.; Urbach, Y.K.; Schulze-Krebs, A.; Canneva, F.; Moceri, S.; Habermeyer, J.; Achoui, D.; Gupta, B.; Steindler, D.A.; et al. Early postnatal behavioral, cellular, and molecular changes in models of Huntington disease are reversible by HDAC inhibition. Proc. Natl. Acad. Sci. USA 2018, 115, E8765–E8774. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Chopra, V.; Quinti, L.; Khanna, P.; Paganetti, P.; Kuhn, R.; Young, A.B.; Kazantsev, A.G.; Hersch, S. LBH589, A Hydroxamic Acid-Derived HDAC Inhibitor, is Neuroprotective in Mouse Models of Huntington’s Disease. J. Huntingtons. Dis. 2016, 5, 347–355. [Google Scholar] [CrossRef] [Green Version]
  96. Jia, H.; Morris, C.D.; Williams, R.M.; Loring, J.F.; Thomas, E.A. HDAC inhibition imparts beneficial transgenerational effects in Huntington’s disease mice via altered DNA and histone methylation. Proc. Natl. Acad. Sci. USA 2014, 112, E56–E64. [Google Scholar] [CrossRef] [Green Version]
  97. Naia, L.; Cunha-Oliveira, T.; Rodrigues, J.; Rosenstock, T.R.; Oliveira, A.; Ribeiro, M.; Carmo, C.; Oliveira-Sousa, S.I.; Duarte, A.I.; Hayden, M.R.; et al. Histone Deacetylase Inhibitors Protect Against Pyruvate Dehydrogenase Dysfunction in Huntington’s Disease. J. Neurosci. 2017, 37, 2776–2794. [Google Scholar] [CrossRef] [Green Version]
  98. Federspiel, J.D.; Greco, T.M.; Lum, K.K.; Cristea, I.M. Hdac4 Interactions in Huntington’s Disease Viewed Through the Prism of Multiomics. Mol. Cell. Proteom. 2019, 18, S92–S113. [Google Scholar] [CrossRef] [Green Version]
  99. Moreno, C.L.; Ehrlich, M.E.; Mobbs, C.V. Protection by dietary restriction in the YAC128 mouse model of Huntington’s disease: Relation to genes regulating histone acetylation and HTT. Neurobiol. Dis. 2016, 85, 25–34. [Google Scholar] [CrossRef] [Green Version]
  100. Jia, H.; Wang, Y.; Morris, C.D.; Jacques, V.; Gottesfeld, J.; Rusche, J.R.; Thomas, E.A. The Effects of Pharmacological Inhibition of Histone Deacetylase 3 (HDAC3) in Huntington’s Disease Mice. PLoS ONE 2016, 11, e0152498. [Google Scholar] [CrossRef]
  101. Suelves, N.; Kirkham-McCarthy, L.; Lahue, R.S.; Ginés, S. A selective inhibitor of histone deacetylase 3 prevents cognitive deficits and suppresses striatal CAG repeat expansions in Huntington’s disease mice. Sci. Rep. 2017, 7, 6082. [Google Scholar] [CrossRef] [Green Version]
  102. Ragot, A.; Pietropaolo, S.; Vincent, J.; Delage, P.; Zhang, H.; Allinquant, B.; Leinekugel, X.; Fischer, A.; Cho, Y.H. Genetic deletion of the Histone Deacetylase 6 exacerbates selected behavioral deficits in the R6/1 mouse model for Huntington’s disease. Brain Behav. 2015, 5, e00361. [Google Scholar] [CrossRef] [PubMed]
  103. Dong, X.; Tsuji, J.; Labadorf, A.; Roussos, P.; Chen, J.-F.; Myers, R.H.; Akbarian, S.; Weng, Z. The Role of H3K4me3 in Transcriptional Regulation Is Altered in Huntington’s Disease. PLoS ONE 2015, 10, e0144398. [Google Scholar] [CrossRef]
  104. Song, W.; Zsindely, N.; Faragó, A.; Marsh, J.L.; Bodai, L. Systematic genetic interaction studies identify histone demethylase Utx as potential target for ameliorating Huntington’s disease. Hum. Mol. Genet. 2017, 27, 649–666. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Lee, J.; Hwang, Y.J.; Kim, Y.; Lee, M.Y.; Hyeon, S.J.; Lee, S.; Kim, D.H.; Jang, S.; Im, H.; Min, S.-J.; et al. Remodeling of heterochromatin structure slows neuropathological progression and prolongs survival in an animal model of Huntington’s disease. Acta Neuropathol. 2017, 134, 729–748. [Google Scholar] [CrossRef] [PubMed]
  106. March, Z.M.; King, O.D.; Shorter, J. Prion-like domains as epigenetic regulators, scaffolds for subcellular organization, and drivers of neurodegenerative disease. Brain Res. 2016, 1647, 9–18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Chen-Plotkin, A.S.; Lee, V.M.-Y.; Trojanowski, J.Q. TAR DNA-binding protein 43 in neurodegenerative disease. Nat. Rev. Neurol. 2010, 6, 211–220. [Google Scholar] [CrossRef] [Green Version]
  108. Guo, L.; Shorter, J. Biology and Pathobiology of TDP-43 and Emergent Therapeutic Strategies. Cold Spring Harb. Perspect. Med. 2016, 7, a024554. [Google Scholar] [CrossRef] [Green Version]
  109. Harrison, A.F.; Shorter, J. RNA-binding proteins with prion-like domains in health and disease. Biochem. J. 2017, 474, 1417–1438. [Google Scholar] [CrossRef] [Green Version]
  110. Murakami, K.; Ono, K. Interactions of amyloid coaggregates with biomolecules and its relevance to neurodegeneration. FASEB J. 2022, 36, e22493. [Google Scholar] [CrossRef]
  111. Henriques, A.D.; Machado-Silva, W.; Leite, R.E.P.; Suemoto, C.K.; Leite, K.R.M.; Srougi, M.; Pereira, A.C.; Jacob-Filho, W.; Nóbrega, O.T. Genome-wide profiling and predicted significance of post-mortem brain microRNA in Alzheimer’s disease. Mech. Ageing Dev. 2020, 191, 111352. [Google Scholar] [CrossRef] [PubMed]
  112. Moradifard, S.; Hoseinbeyki, M.; Ganji, S.M.; Minuchehr, Z. Analysis of microRNA and Gene Expression Profiles in Alzheimer’s Disease: A Meta-Analysis Approach. Sci. Rep. 2018, 8, 4767. [Google Scholar] [CrossRef] [PubMed]
  113. Nunez-Iglesias, J.; Liu, C.-C.; Morgan, T.E.; Finch, C.E.; Zhou, X.J. Joint Genome-Wide Profiling of miRNA and mRNA Expression in Alzheimer’s Disease Cortex Reveals Altered miRNA Regulation. PLoS ONE 2010, 5, e8898. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Watson, C.N.; Begum, G.; Ashman, E.; Thorn, D.; Yakoub, K.M.; Al Hariri, M.; Nehme, A.; Mondello, S.; Kobeissy, F.; Belli, A.; et al. Co-Expression Analysis of microRNAs and Proteins in Brain of Alzheimer’s Disease Patients. Cells 2022, 11, 163. [Google Scholar] [CrossRef] [PubMed]
  115. Ren, R.J.; Zhang, Y.F.; Dammer, E.B.; Zhou, Y.; Wang, L.L.; Liu, X.H.; Feng, B.L.; Jiang, G.X.; Chen, S.D.; Wang, G.; et al. Peripheral Blood MicroRNA Expression Profiles in Alzheimer’s Disease: Screening, Validation, Association with Clinical Phenotype and Implications for Molecular Mechanism. Mol. Neurobiol. 2016, 53, 5772–5781. [Google Scholar] [CrossRef]
  116. Wang, Z.; Shen, L.; Wang, Y.; Huang, S. Integrated analysis of miRNA and mRNA expression in the blood of patients with Alzheimer’s disease. Mol. Med. Rep. 2020, 22, 1053–1062. [Google Scholar] [CrossRef]
  117. Dong, H.; Li, J.; Huang, L.; Chen, X.; Li, D.; Wang, T.; Hu, C.; Xu, J.; Zhang, C.; Zen, K.; et al. Serum MicroRNA Profiles Serve as Novel Biomarkers for the Diagnosis of Alzheimer’s Disease. Dis. Markers 2015, 2015, 625659. [Google Scholar] [CrossRef] [Green Version]
  118. Lu, L.; Dai, W.-Z.; Zhu, X.-C.; Ma, T. Analysis of Serum miRNAs in Alzheimer’s Disease. Am. J. Alzheimer’s Dis. Other Dementias 2021, 36. [Google Scholar] [CrossRef]
  119. Dangla-Valls, A.; Molinuevo, J.L.; Altirriba, J.; Sánchez-Valle, R.; Alcolea, D.; Fortea, J.; Rami, L.; Balasa, M.; Muñoz-García, C.; Ezquerra, M.; et al. CSF microRNA Profiling in Alzheimer’s Disease: A Screening and Validation Study. Mol. Neurobiol. 2016, 54, 6647–6654. [Google Scholar] [CrossRef]
  120. Dong, Z.; Gu, H.; Guo, Q.; Liang, S.; Xue, J.; Yao, F.; Liu, X.; Li, F.; Liu, H.; Sun, L.; et al. Profiling of Serum Exosome MiRNA Reveals the Potential of a MiRNA Panel as Diagnostic Biomarker for Alzheimer’s Disease. Mol. Neurobiol. 2021, 58, 3084–3094. [Google Scholar] [CrossRef]
  121. McKeever, P.M.; Schneider, R.; Taghdiri, F.; Weichert, A.; Multani, N.; Brown, R.A.; Boxer, A.L.; Karydas, A.; Miller, B.; Robertson, J.; et al. MicroRNA Expression Levels Are Altered in the Cerebrospinal Fluid of Patients with Young-Onset Alzheimer’s Disease. Mol. Neurobiol. 2018, 55, 8826–8841. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Serpente, M.; Fenoglio, C.; D’Anca, M.; Arcaro, M.; Sorrentino, F.; Visconte, C.; Arighi, A.; Fumagalli, G.G.; Porretti, L.; Cattaneo, A.; et al. MiRNA Profiling in Plasma Neural-Derived Small Extracellular Vesicles from Patients with Alzheimer’s Disease. Cells 2020, 9, 1443. [Google Scholar] [CrossRef] [PubMed]
  123. Ma, N.; Tie, C.; Yu, B.; Zhang, W.; Wan, J. Identifying lncRNA-miRNA-mRNA networks to investigate Alzheimer’s disease pathogenesis and therapy strategy. Aging 2020, 12, 2897–2920. [Google Scholar] [CrossRef] [PubMed]
  124. Song, Z.; Qu, Y.; Xu, Y.; Zhang, L.; Zhou, L.; Han, Y.; Zhao, W.; Yu, P.; Zhang, Y.; Li, X.; et al. Microarray microRNA profiling of urinary exosomes in a 5XFAD mouse model of Alzheimer’s disease. Anim. Model. Exp. Med. 2021, 4, 233–242. [Google Scholar] [CrossRef]
  125. Li, Z.-R.; Liu, R.; Zeng, L.; Jiang, H.-L.; Ashraf, G.M. MicroRNA and mRNA profiling of cerebral cortex in a transgenic mouse model of Alzheimer’s disease by RNA sequencing. Neural Regen. Res. 2021, 16, 2099–2108. [Google Scholar] [CrossRef]
  126. Cardo, L.F.; Coto, E.; Ribacoba, R.; Menéndez, M.; Moris, G.; Suárez, E.; Alvarez, V. MiRNA Profile in the Substantia Nigra of Parkinson’s Disease and Healthy Subjects. J. Mol. Neurosci. 2014, 54, 830–836. [Google Scholar] [CrossRef]
  127. Hoss, A.G.; Labadorf, A.; Beach, T.G.; Latourelle, J.C.; Myers, R.H. microRNA Profiles in Parkinson’s Disease Pre-frontal Cortex. Front. Aging Neurosci. 2016, 8, 36. [Google Scholar] [CrossRef] [Green Version]
  128. Schulz, J.; Takousis, P.; Wohlers, I.; Itua, I.O.; Dobricic, V.; Rücker, G.; Binder, H.; Middleton, L.; Ioannidis, J.P.; Perneczky, R.; et al. Meta-analyses identify differentially expressed microRNAs in Parkinson’s disease. Ann. Neurol. 2019, 85, 835–851. [Google Scholar] [CrossRef]
  129. Tatura, R.; Kraus, T.; Giese, A.; Arzberger, T.; Buchholz, M.; Höglinger, G.; Müller, U. Parkinson’s disease: SNCA-, PARK2-, and LRRK2- targeting microRNAs elevated in cingulate gyrus. Park. Relat. Disord. 2016, 33, 115–121. [Google Scholar] [CrossRef]
  130. Kurz, A.; Kumar, R.; Northoff, B.H.; Wenk, C.; Schirra, J.; Donakonda, S.; Höglinger, G.U.; Schwarz, J.; Rozanski, V.; Hübner, R.; et al. Differential expression of gut miRNAs in idiopathic Parkinson’s disease. Park. Relat. Disord. 2021, 88, 46–50. [Google Scholar] [CrossRef]
  131. Cai, M.; Chai, S.; Xiong, T.; Wei, J.; Mao, W.; Zhu, Y.; Li, X.; Wei, W.; Dai, X.; Yang, B.; et al. Aberrant Expression of Circulating MicroRNA Leads to the Dysregulation of Alpha-Synuclein and Other Pathogenic Genes in Parkinson’s Disease. Front. Cell Dev. Biol. 2021, 9, 695007. [Google Scholar] [CrossRef]
  132. Cardo, L.F.; Coto, E.; de Mena, L.; Ribacoba, R.; Moris, G.; Menéndez, M.; Álvarez, L.D.M. Profile of microRNAs in the plasma of Parkinson’s disease patients and healthy controls. J. Neurol. 2013, 260, 1420–1422. [Google Scholar] [CrossRef] [PubMed]
  133. Ruf, W.; Freischmidt, A.; Grozdanov, V.; Roth, V.; Brockmann, S.; Mollenhauer, B.; Martin, D.; Haslinger, B.; Fundel-Clemens, K.; Otto, M.; et al. Protein Binding Partners of Dysregulated miRNAs in Parkinson’s Disease Serum. Cells 2021, 10, 791. [Google Scholar] [CrossRef] [PubMed]
  134. dos Santos, M.C.T.; Barreto-Sanz, M.A.; Correia, B.R.S.; Bell, R.; Widnall, C.; Perez, L.T.; Berteau, C.; Schulte, C.; Scheller, D.; Berg, D.; et al. miRNA-based signatures in cerebrospinal fluid as potential diagnostic tools for early stage Parkinson’s disease. Oncotarget 2018, 9, 17455–17465. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Mo, M.; Xiao, Y.; Huang, S.; Cen, L.; Chen, X.; Zhang, L.; Luo, Q.; Li, S.; Yang, X.; Lin, X.; et al. MicroRNA expressing profiles in A53T mutant alpha-synuclein transgenic mice and Parkinsonian. Oncotarget 2016, 8, 15–28. [Google Scholar] [CrossRef] [Green Version]
  136. Jiang, Y.; Chen, J.; Sun, Y.; Li, F.; Wei, L.; Sun, W.; Deng, J.; Yuan, Y.; Wang, Z. Profiling of Differentially Expressed MicroRNAs in Saliva of Parkinson’s Disease Patients. Front. Neurol. 2021, 12, 738530. [Google Scholar] [CrossRef]
  137. He, S.; Huang, L.; Shao, C.; Nie, T.; Xia, L.; Cui, B.; Lu, F.; Zhu, L.; Chen, B.; Yang, Q. Several miRNAs derived from serum extracellular vesicles are potential biomarkers for early diagnosis and progression of Parkinson’s disease. Transl. Neurodegener. 2021, 10, 25. [Google Scholar] [CrossRef]
  138. Yadav, S.K.; Pandey, A.; Sarkar, S.; Yadav, S.S.; Parmar, D.; Yadav, S. Identification of Altered Blood MicroRNAs and Plasma Proteins in a Rat Model of Parkinson’s Disease. Mol. Neurobiol. 2022, 59, 1781–1798. [Google Scholar] [CrossRef]
  139. Tolosa, E.; Botta-Orfila, T.; Morató, X.; Calatayud, C.; Ferrer-Lorente, R.; Martí, M.J.; Fernández, M.; Gaig, C.; Raya, A.; Consiglio, A.; et al. MicroRNA alterations in iPSC-derived dopaminergic neurons from Parkinson disease patients. Neurobiol. Aging 2018, 69, 283–291. [Google Scholar] [CrossRef]
  140. Campos-Melo, D.; Droppelmann, C.A.; He, Z.; Volkening, K.; Strong, M.J. Altered microRNA expression profile in amyotrophic lateral sclerosis: A role in the regulation of NFL mRNA levels. Mol. Brain 2013, 6, 26. [Google Scholar] [CrossRef] [Green Version]
  141. Wakabayashi, K.; Mori, F.; Kakita, A.; Takahashi, H.; Utsumi, J.; Sasaki, H. Analysis of microRNA from archived formalin-fixed paraffin-embedded specimens of amyotrophic lateral sclerosis. Acta Neuropathol. Commun. 2014, 2, 173. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Aksu-Menges, E.; Balci-Hayta, B.; Bekircan-Kurt, C.E.; Aydinoglu, A.T.; Erdem-Ozdamar, S.; Tan, E. Two distinct skeletal muscle microRNA signatures revealing the complex mechanism of sporadic ALS. Acta Neurol. Belg. 2021, 1–11. [Google Scholar] [CrossRef] [PubMed]
  143. De Felice, B.; Manfellotto, F.; Fiorentino, G.; Annunziata, A.; Biffali, E.; Pannone, R.; Federico, A. Wide-Ranging Analysis of MicroRNA Profiles in Sporadic Amyotrophic Lateral Sclerosis Using Next-Generation Sequencing. Front. Genet. 2018, 9, 310. [Google Scholar] [CrossRef] [PubMed]
  144. Chen, Y.; Wei, Q.; Chen, X.; Li, C.; Cao, B.; Ou, R.; Hadano, S.; Shang, H.-F. Aberration of miRNAs Expression in Leukocytes from Sporadic Amyotrophic Lateral Sclerosis. Front. Mol. Neurosci. 2016, 9, 69. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. De Felice, B.; Guida, M.; Guida, M.; Coppola, C.; de Mieri, G.; Cotrufo, R. A miRNA signature in leukocytes from sporadic amyotrophic lateral sclerosis. Gene 2012, 508, 35–40. [Google Scholar] [CrossRef]
  146. Liguori, M.; Nuzziello, N.; Introna, A.; Consiglio, A.; Licciulli, F.; D’Errico, E.; Scarafino, A.; Distaso, E.; Simone, I.L. Dysregulation of MicroRNAs and Target Genes Networks in Peripheral Blood of Patients With Sporadic Amyotrophic Lateral Sclerosis. Front. Mol. Neurosci. 2018, 11, 288. [Google Scholar] [CrossRef]
  147. Takahashi, I.; Hama, Y.; Matsushima, M.; Hirotani, M.; Kano, T.; Hohzen, H.; Yabe, I.; Utsumi, J.; Sasaki, H. Identification of plasma microRNAs as a biomarker of sporadic Amyotrophic Lateral Sclerosis. Mol. Brain 2015, 8, 67. [Google Scholar] [CrossRef]
  148. Freischmidt, A.; Müller, K.; Zondler, L.; Weydt, P.; Volk, A.E.; Božič, A.L.; Walter, M.; Bonin, M.; Mayer, B.; von Arnim, C.A.F.; et al. Serum microRNAs in patients with genetic amyotrophic lateral sclerosis and pre-manifest mutation carriers. Brain 2014, 137, 2938–2950. [Google Scholar] [CrossRef] [Green Version]
  149. Joilin, G.; Gray, E.; Thompson, A.G.; Bobeva, Y.; Talbot, K.; Weishaupt, J.; Ludolph, A.; Malaspina, A.; Leigh, P.N.; Newbury, S.F.; et al. Identification of a potential non-coding RNA biomarker signature for amyotrophic lateral sclerosis. Brain Commun. 2020, 2, fcaa053. [Google Scholar] [CrossRef]
  150. Taguchi, Y.-H.; Wang, H. Exploring microRNA Biomarker for Amyotrophic Lateral Sclerosis. Int. J. Mol. Sci. 2018, 19, 1318. [Google Scholar] [CrossRef] [Green Version]
  151. Benigni, M.; Ricci, C.; Jones, A.R.; Giannini, F.; Al-Chalabi, A.; Battistini, S. Identification of miRNAs as Potential Biomarkers in Cerebrospinal Fluid from Amyotrophic Lateral Sclerosis Patients. NeuroMolecular Med. 2016, 18, 551–560. [Google Scholar] [CrossRef] [PubMed]
  152. Katsu, M.; Hama, Y.; Utsumi, J.; Takashina, K.; Yasumatsu, H.; Mori, F.; Wakabayashi, K.; Shoji, M.; Sasaki, H. MicroRNA expression profiles of neuron-derived extracellular vesicles in plasma from patients with am-yotrophic lateral sclerosis. Neurosci. Lett. 2019, 708, 134176. [Google Scholar] [CrossRef] [PubMed]
  153. Lo, T.-W.; Figueroa-Romero, C.; Hur, J.; Pacut, C.; Stoll, E.; Spring, C.; Lewis, R.; Nair, A.; Goutman, S.A.; Sakowski, S.A.; et al. Extracellular Vesicles in Serum and Central Nervous System Tissues Contain microRNA Signatures in Sporadic Amyotrophic Lateral Sclerosis. Front. Mol. Neurosci. 2021, 14, 739016. [Google Scholar] [CrossRef] [PubMed]
  154. Rizzuti, M.; Melzi, V.; Gagliardi, D.; Resnati, D.; Meneri, M.; Dioni, L.; Masrori, P.; Hersmus, N.; Poesen, K.; Locatelli, M.; et al. Insights into the identification of a molecular signature for amyotrophic lateral sclerosis exploiting integrated microRNA profiling of iPSC-derived motor neurons and exosomes. Cell. Mol. Life Sci. 2022, 79, 189. [Google Scholar] [CrossRef]
  155. Saucier, D.; Wajnberg, G.; Roy, J.; Beauregard, A.P.; Chacko, S.; Crapoulet, N.; Fournier, S.; Ghosh, A.; Lewis, S.M.; Marrero, A.; et al. Identification of a circulating miRNA signature in extracellular vesicles collected from amyotrophic lateral sclerosis patients. Brain Res. 2019, 1708, 100–108. [Google Scholar] [CrossRef]
  156. Toivonen, J.M.; Manzano, R.; Oliván, S.; Zaragoza, P.; García-Redondo, A.; Osta, R. MicroRNA-206: A Potential Circulating Biomarker Candidate for Amyotrophic Lateral Sclerosis. PLoS ONE 2014, 9, e89065. [Google Scholar] [CrossRef] [Green Version]
  157. Matamala, J.M.; Arias-Carrasco, R.; Sanchez, C.; Uhrig, M.; Bargsted, L.; Matus, S.; Maracaja-Coutinho, V.; Abarzua, S.; van Zundert, B.; Verdugo, R.; et al. Genome-wide circulating microRNA expression profiling reveals potential biomarkers for amyotrophic lateral sclerosis. Neurobiol. Aging 2018, 64, 123–138. [Google Scholar] [CrossRef]
  158. Rizzuti, M.; Filosa, G.; Melzi, V.; Calandriello, L.; Dioni, L.; Bollati, V.; Bresolin, N.; Comi, G.P.; Barabino, S.; Nizzardo, M.; et al. MicroRNA expression analysis identifies a subset of downregulated miRNAs in ALS motor neuron progenitors. Sci. Rep. 2018, 8, 10105. [Google Scholar] [CrossRef] [Green Version]
  159. Dong, X.; Cong, S. Bioinformatic analysis of microRNA expression in Huntington’s disease. Mol. Med. Rep. 2018, 18, 2857–2865. [Google Scholar] [CrossRef]
  160. Hoss, A.G.; Kartha, V.K.; Dong, X.; Latourelle, J.C.; Dumitriu, A.; Hadzi, T.C.; Macdonald, M.E.; Gusella, J.F.; Akbarian, S.; Chen, J.-F.; et al. MicroRNAs Located in the Hox Gene Clusters Are Implicated in Huntington’s Disease Pathogenesis. PLoS Genet. 2014, 10, e1004188. [Google Scholar] [CrossRef]
  161. Wang, Z.M.; Dong, X.Y.; Cong, S.Y. Bioinformatic analysis of a microRNA regulatory network in Huntington’s disease. J. Integr. Neurosci. 2020, 19, 641–650. [Google Scholar] [CrossRef] [PubMed]
  162. Díez-Planelles, C.; Sánchez-Lozano, P.; Crespo, M.; Gil-Zamorano, J.; Ribacoba, R.; González, N.; Suárez, E.; Martínez-Descals, A.; Martínez-Camblor, P.; Álvarez, V.; et al. Circulating microRNAs in Huntington’s disease: Emerging mediators in metabolic impairment. Pharmacol. Res. 2016, 108, 102–110. [Google Scholar] [CrossRef] [PubMed]
  163. Reed, E.R.; Latourelle, J.C.; Bockholt, J.H.; Bregu, J.; Smock, J.; Paulsen, J.S.; Myers, R.H. PREDICT-HD CSF ancillary study investigators MicroRNAs in CSF as prodromal biomarkers for Huntington disease in the PREDICT-HD study. Neurology 2017, 90, e264–e272. [Google Scholar] [CrossRef] [PubMed]
  164. Dubois, C.; Kong, G.; Tran, H.; Li, S.; Pang, T.Y.; Hannan, A.J.; Renoir, T. Small Non-coding RNAs Are Dysregulated in Huntington’s Disease Transgenic Mice Independently of the Therapeutic Effects of an Environmental Intervention. Mol. Neurobiol. 2021, 58, 3308–3318. [Google Scholar] [CrossRef]
  165. Langfelder, P.; Gao, F.; Wang, N.; Howland, D.; Kwak, S.; Vogt, T.F.; Aaronson, J.S.; Rosinski, J.; Coppola, G.; Horvath, S.; et al. MicroRNA signatures of endogenous Huntingtin CAG repeat expansion in mice. PLoS ONE 2018, 13, e0190550. [Google Scholar] [CrossRef] [Green Version]
  166. Olmo, I.G.; Olmo, R.P.; Gonçalves, A.N.A.; Pires, R.G.W.; Marques, J.T.; Ribeiro, F.M. High-Throughput Sequencing of BACHD Mice Reveals Upregulation of Neuroprotective miRNAs at the Pre-Symptomatic Stage of Huntington’s Disease. ASN Neuro 2021, 13. [Google Scholar] [CrossRef]
  167. Pircs, K.; Petri, R.; Madsen, S.; Brattås, P.L.; Vuono, R.; Ottosson, D.R.; St-Amour, I.; Hersbach, B.; Matusiak-Brückner, M.; Lundh, S.H.; et al. Huntingtin Aggregation Impairs Autophagy, Leading to Argonaute-2 Accumulation and Global MicroRNA Dysregulation. Cell Rep. 2018, 24, 1397–1406. [Google Scholar] [CrossRef] [Green Version]
  168. Li, K.; Zhang, J.; Ji, C.; Wang, L. MiR-144-3p and Its Target Gene β-Amyloid Precursor Protein Regulate 1-Methyl-4-Phenyl-1,2-3,6-Tetrahydropyridine-Induced Mitochondrial Dysfunction. Mol. Cells 2016, 39, 543–549. [Google Scholar] [CrossRef] [Green Version]
  169. Jovicic, A.; Zaldivar Jolissaint, J.F.; Moser, R.; de Fatima Silva Santos, M.; Luthi-Carter, R. MicroRNA-22 (miR-22) Overexpression Is Neuroprotective via General Anti-Apoptotic Effects and May also Target Specific Huntington’s Disease-Related Mechanisms. PLoS ONE 2013, 8, e54222. [Google Scholar]
  170. Cao, F.; Liu, Z.; Sun, G. Diagnostic value of miR-193a-3p in Alzheimer’s disease and miR-193a-3p attenuates amyloid-β induced neurotoxicity by targeting PTEN. Exp. Gerontol. 2019, 130, 110814. [Google Scholar] [CrossRef]
  171. Yang, Q.; Zhao, Q.; Yin, Y. miR-133b is a potential diagnostic biomarker for Alzheimer’s disease and has a neuroprotective role. Exp. Ther. Med. 2019, 18, 2711–2718. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  172. Wang, D.; Fei, Z.; Luo, S.; Wang, H. MiR-335-5p Inhibits β-Amyloid (Aβ) Accumulation to Attenuate Cognitive Deficits Through Targeting c-jun-N-terminal Kinase 3 in Alzheimer’s Disease. Curr. Neurovasc. Res. 2020, 17, 93–101. [Google Scholar] [CrossRef] [PubMed]
  173. Ji, Y.; Wang, D.; Zhang, B.; Lu, H. MiR-361-3p inhibits β-amyloid accumulation and attenuates cognitive deficits through targeting BACE1 in Alzheimer’s disease. J. Integr. Neurosci. 2019, 18, 285–291. [Google Scholar] [CrossRef] [PubMed]
  174. Wang, L.; Liu, J.; Wang, Q.; Jiang, H.; Zeng, L.; Li, Z.; Liu, R. MicroRNA-200a-3p Mediates Neuroprotection in Alzheimer-Related Deficits and Attenuates Amyloid-Beta Overproduction and Tau Hyperphosphorylation via Coregulating BACE1 and PRKACB. Front. Pharmacol. 2019, 10, 806. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  175. Xu, W.; Li, K.; Fan, Q.; Zong, B.; Han, L. Knockdown of long non-coding RNA SOX21-AS1 attenuates amyloid-β-induced neuronal damage by sponging miR-107. Biosci. Rep. 2020, 40, BSR20194295. [Google Scholar] [CrossRef] [Green Version]
  176. Tan, X.; Luo, Y.; Pi, D.; Xia, L.; Li, Z.; Tu, Q. miR-340 reduces accumulation of amyloid-β through targeting BACE1 (β-site amyloid precursor protein cleaving enzyme 1) in Alzheimer’ s disease. Curr. Neurovasc. Res. 2020, 17, 86–92. [Google Scholar] [CrossRef]
  177. Baby, N.; Alagappan, N.; Dheen, S.T.; Sajikumar, S. MicroRNA-134-5p inhibition rescues long-term plasticity and synaptic tagging/capture in an Aβ(1–42)-induced model of Alzheimer’s disease. Aging Cell 2020, 19, e13046. [Google Scholar] [CrossRef] [Green Version]
  178. Hou, T.Y.; Zhou, Y.; Zhu, L.S.; Wang, X.; Pang, P.; Wang, D.Q.; Liuyang, Z.Y.; Man, H.; Lu, Y.; Zhu, L.Q.; et al. Correcting abnormalities in miR-124/PTPN1 signaling rescues tau pathology in Alzheimer’s disease. J. Neurochem. 2020, 154, 441–457. [Google Scholar] [CrossRef]
  179. Shi, Z.; Zhang, K.; Zhou, H.; Jiang, L.; Xie, B.; Wang, R.; Xia, W.; Yin, Y.; Gao, Z.; Cui, D.; et al. Increased miR-34c mediates synaptic deficits by targeting synaptotagmin 1 through ROS-JNK-p53 pathway in Alzheimer’s Disease. Aging Cell 2020, 19, e13125. [Google Scholar] [CrossRef] [Green Version]
  180. Wang, X.; Liu, D.; Huang, H.Z.; Wang, Z.H.; Hou, T.Y.; Yang, X.; Pang, P.; Wei, N.; Zhou, Y.F.; Dupras, M.J.; et al. A Novel MicroRNA-124/PTPN1 Signal Pathway Mediates Synaptic and Memory Deficits in Alzheimer’s Disease. Biol. Psychiatry 2018, 83, 395–405. [Google Scholar] [CrossRef]
  181. Ansari, A.; Maffioletti, E.; Milanesi, E.; Marizzoni, M.; Frisoni, G.B.; Blin, O.; Richardson, J.C.; Bordet, R.; Forloni, G.; Gennarelli, M.; et al. miR-146a and miR-181a are involved in the progression of mild cognitive impairment to Alzheimer’s disease. Neurobiol. Aging 2019, 82, 102–109. [Google Scholar] [CrossRef] [PubMed]
  182. Ghanbari, M.; Munshi, S.T.; Ma, B.; Lendemeijer, B.; Bansal, S.; Adams, H.H.; Wang, W.; Goth, K.; Slump, D.E.; van den Hout, M.C.G.N.; et al. A functional variant in the miR-142 promoter modulating its expression and conferring risk of Alzheimer disease. Hum. Mutat. 2019, 40, 2131–2145. [Google Scholar] [CrossRef] [PubMed]
  183. Li, H.; Yu, L.; Li, M.; Chen, X.; Tian, Q.; Jiang, Y.; Li, N. MicroRNA-150 serves as a diagnostic biomarker and is involved in the inflammatory pathogenesis of Parkinson’s disease. Mol. Genet. Genom. Med. 2020, 8, e1189. [Google Scholar] [CrossRef]
  184. Zhang, J.; Zhou, D.; Zhang, Z.; Qu, X.; Bao, K.; Lu, G.; Duan, J. miR-let-7a suppresses α-Synuclein-induced microglia inflammation through targeting STAT3 in Parkinson’s disease. Biochem. Biophys. Res. Commun. 2019, 519, 740–746. [Google Scholar] [CrossRef] [PubMed]
  185. Sun, Q.; Wang, S.; Chen, J.; Cai, H.; Huang, W.; Zhang, Y.; Wang, L.; Xing, Y. MicroRNA-190 alleviates neuronal damage and inhibits neuroinflammation via Nlrp3 in MPTP-induced Parkinson’s disease mouse model. J. Cell. Physiol. 2019, 234, 23379–23387. [Google Scholar] [CrossRef] [PubMed]
  186. Zeng, R.; Luo, D.-X.; Li, H.-P.; Zhang, Q.-S.; Lei, S.-S.; Chen, J.-H. MicroRNA-135b alleviates MPP+-mediated Parkinson’s disease in in vitro model through suppressing FoxO1-induced NLRP3 inflammasome and pyroptosis. J. Clin. Neurosci. 2019, 65, 125–133. [Google Scholar] [CrossRef] [PubMed]
  187. Cai, L.J.; Tu, L.; Li, T.; Yang, X.L.; Ren, Y.P.; Gu, R.; Zhang, Q.; Yao, H.; Qu, X.; Wang, Q.; et al. Up-regulation of microRNA-375 ameliorates the damage of dopaminergic neurons, reduces oxidative stress and inflammation in Parkinson’s disease by inhibiting SP1. Aging 2020, 12, 672–689. [Google Scholar] [CrossRef]
  188. Wang, R.; Yang, Y.; Wang, H.; He, Y.; Li, C. MiR-29c protects against inflammation and apoptosis in Parkinson’s disease model in vivo and in vitro by targeting SP1. Clin. Exp. Pharmacol. Physiol. 2019, 47, 372–382. [Google Scholar] [CrossRef]
  189. Liu, W.; Li, L.; Liu, S.; Wang, Z.; Kuang, H.; Xia, Y.; Tang, C.; Yin, D. MicroRNA Expression Profiling Screen miR-3557/324-Targeted CaMK/mTOR in the Rat Striatum of Parkinson’s Disease in Regular Aerobic Exercise. Biomed Res. Int. 2019, 2019, 7654798. [Google Scholar] [CrossRef] [Green Version]
  190. Ge, H.; Yan, Z.; Zhu, H.; Zhao, H. MiR-410 exerts neuroprotective effects in a cellular model of Parkinson’s disease induced by 6-hydroxydopamine via inhibiting the PTEN/AKT/mTOR signaling pathway. Exp. Mol. Pathol. 2019, 109, 16–24. [Google Scholar] [CrossRef]
  191. Zhao, X.H.; Wang, Y.B.; Yang, J.; Liu, H.Q.; Wang, L.L. MicroRNA-326 suppresses iNOS expression and promotes autophagy of dopaminergic neurons through the JNK signaling by targeting XBP1 in a mouse model of Parkinson’s disease. J. Cell. Biochem. 2019, 120, 14995–15006. [Google Scholar] [CrossRef] [PubMed]
  192. Ren, Y.; Li, H.; Xie, W.; Wei, N.; Liu, M. MicroRNA-195 triggers neuroinflammation in Parkinson’s disease in a Rho-associated kinase 1-dependent manner. Mol. Med. Rep. 2019, 19, 5153–5161. [Google Scholar] [CrossRef] [PubMed]
  193. Li, B.; Jiang, Y.; Xu, Y.; Li, Y.; Li, B. Identification of miRNA-7 as a regulator of brain-derived neurotrophic fac-tor/A-synuclein axis in atrazine-induced Parkinson’s disease by peripheral blood and brain microRNA profiling. Chemosphere 2019, 233, 542–548. [Google Scholar] [CrossRef] [PubMed]
  194. Baghi, M.; Delavar, M.R.; Yadegari, E.; Peymani, M.; Pozo, D.; Nasr-Esfahani, M.H.; Ghaedi, K. Modified level of miR-376a is associated with Parkinson’s disease. J. Cell. Mol. Med. 2020, 24, 2622–2634. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  195. De Luna, N.; Turon-Sans, J.; Cortes-Vicente, E.; Carrasco-Rozas, A.; Illán-Gala, I.; Dols-Icardo, O.; Clarimón, J.; Lleó, A.; Gallardo, E.; Illa, I.; et al. Downregulation of miR-335-5P in Amyotrophic Lateral Sclerosis Can Contribute to Neuronal Mitochondrial Dysfunction and Apoptosis. Sci. Rep. 2020, 10, 4308. [Google Scholar] [CrossRef] [Green Version]
  196. Rohm, M.; May, C.; Marcus, K.; Steinbach, S.; Theis, V.; Theiss, C.; Matschke, V. The microRNA miR-375-3p and the tumor suppressor NDRG2 are involved in sporadic amyotrophic lateral sclerosis. Cell. Physiol. Biochem. 2019, 52, 1412–1426. [Google Scholar]
  197. Klatt, C.L.; Theis, V.; Hahn, S.; Theiss, C.; Matschke, V. Deregulated miR-29b-3p Correlates with Tissue-Specific Activation of Intrinsic Apoptosis in An Animal Model of Amyotrophic Lateral Sclerosis. Cells 2019, 8, 1077. [Google Scholar] [CrossRef] [Green Version]
  198. Maimon, R.; Ionescu, A.; Bonnie, A.; Sweetat, S.; Wald-Altman, S.; Inbar, S.; Gradus, T.; Trotti, D.; Weil, M.; Behar, O.; et al. miR126-5p Downregulation Facilitates Axon Degeneration and NMJ Disruption via a Non-Cell-Autonomous Mechanism in ALS. J. Neurosci. 2018, 38, 5478–5494. [Google Scholar] [CrossRef] [Green Version]
  199. Varcianna, A.; Myszczynska, M.A.; Castelli, L.M.; O’Neill, B.; Kim, Y.; Talbot, J.; Nyberg, S.; Nyamali, I.; Heath, P.R.; Stopford, M.J.; et al. Micro-RNAs secreted through astrocyte-derived extracellular vesicles cause neuronal network degeneration in C9orf72 ALS. eBioMedicine 2019, 40, 626–635. [Google Scholar] [CrossRef] [Green Version]
  200. Helferich, A.M.; Brockmann, S.J.; Reinders, J.; Deshpande, D.; Holzmann, K.; Brenner, D.; Andersen, P.M.; Petri, S.; Thal, D.R.; Michaelis, J.; et al. Dysregulation of a novel miR-1825/TBCB/TUBA4A pathway in sporadic and familial ALS. Cell. Mol. Life Sci. 2018, 75, 4301–4319. [Google Scholar] [CrossRef]
  201. Li, C.; Wei, Q.; Gu, X.; Chen, Y.; Chen, X.; Cao, B.; Ou, R.; Shang, H. Decreased Glycogenolysis by miR-338-3p Promotes Regional Glycogen Accumulation Within the Spinal Cord of Amyotrophic Lateral Sclerosis Mice. Front. Mol. Neurosci. 2019, 12, 114. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  202. Hawley, Z.C.E.; Campos-Melo, D.; Strong, M.J. MiR-105 and miR-9 regulate the mRNA stability of neuronal intermediate filaments. Implications for the pathogenesis of amyotrophic lateral sclerosis (ALS). Brain Res. 2019, 1706, 93–100. [Google Scholar] [CrossRef] [PubMed]
  203. Hoye, M.L.; Regan, M.R.; A Jensen, L.; Lake, A.M.; Reddy, L.V.; Vidensky, S.; Richard, J.-P.; Maragakis, N.J.; Rothstein, J.D.; Dougherty, J.D.; et al. Motor neuron-derived microRNAs cause astrocyte dysfunction in amyotrophic lateral sclerosis. Brain 2018, 141, 2561–2575. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  204. Williams, A.H.; Valdez, G.; Moresi, V.; Qi, X.; McAnally, J.; Elliott, J.L.; Bassel-Duby, R.; Sanes, J.R.; Olson, E.N. MicroRNA-206 Delays ALS Progression and Promotes Regeneration of Neuromuscular Synapses in Mice. Science 2009, 326, 1549–1554. [Google Scholar] [CrossRef]
  205. Tung, Y.-T.; Peng, K.-C.; Chen, Y.-C.; Yen, Y.-P.; Chang, M.; Thams, S.; Chen, J.-A. Mir-17~92 Confers Motor Neuron Subtype Differential Resistance to ALS-Associated Degeneration. Cell Stem Cell 2019, 25, 193–209.e7. [Google Scholar] [CrossRef]
  206. Mégret, L.; Nair, S.S.; Dancourt, J.; Aaronson, J.; Rosinski, J.; Neri, C. Combining feature selection and shape analysis uncovers precise rules for miRNA regulation in Huntington’s disease mice. BMC Bioinform. 2020, 21, 75. [Google Scholar] [CrossRef] [Green Version]
  207. Fukuoka, M.; Takahashi, M.; Fujita, H.; Chiyo, T.; Popiel, H.A.; Watanabe, S.; Furuya, H.; Murata, M.; Wada, K.; Okada, T.; et al. Supplemental Treatment for Huntington’s Disease with miR-132 that Is Deficient in Huntington’s Disease Brain. Mol. Ther. Nucleic Acids 2018, 11, 79–90. [Google Scholar] [CrossRef] [Green Version]
  208. Kunkanjanawan, T.; Carter, R.L.; Prucha, M.S.; Yang, J.; Parnpai, R.; Chan, A.W.S. miR-196a Ameliorates Cytotoxicity and Cellular Phenotype in Transgenic Huntington’s Disease Monkey Neural Cells. PLoS ONE 2016, 11, e0162788. [Google Scholar] [CrossRef] [Green Version]
  209. Her, L.-S.; Mao, S.-H.; Chang, C.-Y.; Cheng, P.-H.; Chang, Y.-F.; Yang, H.-I.; Chen, C.-M.; Yang, S.-H. miR-196a Enhances Neuronal Morphology through Suppressing RANBP10 to Provide Neuroprotection in Huntington’s Disease. Theranostics 2017, 7, 2452–2462. [Google Scholar] [CrossRef]
  210. Ban, J.-J.; Chung, J.-Y.; Lee, M.; Im, W.; Kim, M. MicroRNA-27a reduces mutant hutingtin aggregation in an in vitro model of Huntington’s disease. Biochem. Biophys. Res. Commun. 2017, 488, 316–321. [Google Scholar] [CrossRef]
  211. Pfister, E.L.; DiNardo, N.; Mondo, E.; Borel, F.; Conroy, F.; Fraser, C.; Gernoux, G.; Han, X.; Hu, D.; Johnson, E.; et al. Artificial miRNAs Reduce Human Mutant Huntingtin Throughout the Striatum in a Transgenic Sheep Model of Huntington’s Disease. Hum. Gene Ther. 2018, 29, 663–673. [Google Scholar] [CrossRef] [PubMed]
  212. Caron, N.S.; Southwell, A.L.; Brouwers, C.C.; Cengio, L.D.; Xie, Y.; Black, H.F.; Anderson, L.M.; Ko, S.; Zhu, X.; van Deventer, S.J.; et al. Potent and sustained huntingtin lowering via AAV5 encoding miRNA preserves striatal volume and cognitive function in a humanized mouse model of Huntington disease. Nucleic Acids Res. 2019, 48, 36–54. [Google Scholar] [CrossRef] [PubMed]
  213. Cordeiro, Y.; Macedo, B.; Silva, J.L.; Gomes, M.P.B. Pathological implications of nucleic acid interactions with proteins associated with neurodegenerative diseases. Biophys. Rev. 2014, 6, 97–110. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  214. Cordeiro, Y.; Silva, J.L. The Hypothesis of the Catalytic Action of Nucleic Acid on the Conversion of Prion Protein. Protein Pept. Lett. 2005, 12, 251–255. [Google Scholar] [CrossRef]
  215. Macedo, B.; Millen, T.A.; Braga, C.A.C.A.; Gomes, M.P.B.; Ferreira, P.S.; Kraineva, J.; Winter, R.; Silva, J.; Cordeiro, Y. Nonspecific Prion Protein–Nucleic Acid Interactions Lead to Different Aggregates and Cytotoxic Species. Biochemistry 2012, 51, 5402–5413. [Google Scholar] [CrossRef]
  216. Gomes, M.P.B.; Millen, T.A.; Ferreira, P.S.; e Silva, N.L.C.; Vieira, T.C.R.G.; Almeida, M.; Silva, J.L.; Cordeiro, Y. Prion Protein Complexed to N2a Cellular RNAs through Its N-terminal Domain Forms Aggregates and Is Toxic to Murine Neuroblastoma Cells. J. Biol. Chem. 2008, 283, 19616–19625. [Google Scholar] [CrossRef] [Green Version]
  217. Tandon, A.; Subramani, V.K.; Kim, K.K.; Park, S.H. Interaction of Prion Peptides with DNA Structures. ACS Omega 2021, 7, 176–186. [Google Scholar] [CrossRef]
  218. Adler, V.; Zeiler, B.; Kryukov, V.; Kascsak, R.; Rubenstein, R.; Grossman, A. Small, highly structured RNAs participate in the conversion of human recombinant PrPSen to PrPRes in vitro. J. Mol. Biol. 2003, 332, 47–57. [Google Scholar] [CrossRef]
  219. Deleault, N.R.; Lucassen, R.W.; Supattapone, S. RNA molecules stimulate prion protein conversion. Nature 2003, 425, 717–720. [Google Scholar] [CrossRef]
  220. Mashima, T.; Nishikawa, F.; Kamatari, Y.O.; Fujiwara, H.; Saimura, M.; Nagata, T.; Kodaki, T.; Nishikawa, S.; Kuwata, K.; Katahira, M. Anti-prion activity of an RNA aptamer and its structural basis. Nucleic Acids Res. 2012, 41, 1355–1362. [Google Scholar] [CrossRef] [Green Version]
  221. Dalai, W.; Matsuo, E.; Takeyama, N.; Kawano, J.; Saeki, K. CpG site DNA methylation patterns reveal a novel regulatory element in the mouse prion protein gene. J. Vet. Med. Sci. 2017, 79, 100–107. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  222. Dalai, W.; Matsuo, E.; Takeyama, N.; Kawano, J.; Saeki, K. Increased expression of prion protein gene is accompanied by demethylation of CpG sites in a mouse embryonal carcinoma cell line, P19C6. J. Vet. Med. Sci. 2017, 79, 644–648. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  223. Dabin, L.C.; Guntoro, F.; Campbell, T.; Bélicard, T.; Smith, A.R.; Smith, R.G.; Raybould, R.; Schott, J.M.; Lunnon, K.; Sarkies, P.; et al. Altered DNA methylation profiles in blood from patients with sporadic Creutzfeldt–Jakob disease. Acta Neuropathol. 2020, 140, 863–879. [Google Scholar] [CrossRef] [PubMed]
  224. Hernaiz, A.; Sanz, A.; Sentre, S.; Ranera, B.; Lopez-Pérez, O.; Zaragoza, P.; Badiola, J.J.; Filali, H.; Bolea, R.; Toivonen, J.M.; et al. Genome-Wide Methylation Profiling in the Thalamus of Scrapie Sheep. Front. Vet. Sci. 2022, 9, 824677. [Google Scholar] [CrossRef]
  225. Guntoro, F.; Viré, E.; Giordani, C.; Darwent, L.; Hummerich, H.; Linehan, J.; Sinka, K.; Jaunmuktane, Z.; Brandner, S.; Collinge, J.; et al. DNA methylation analysis of archival lymphoreticular tissues in Creutzfeldt–Jakob disease. Acta Neuropathol. 2022, 144, 785–787. [Google Scholar] [CrossRef]
  226. Harvey, Z.H.; Chakravarty, A.K.; Futia, R.A.; Jarosz, D.F. A Prion Epigenetic Switch Establishes an Active Chromatin State. Cell 2020, 180, 928–940.e14. [Google Scholar] [CrossRef]
  227. Zhu, T.; Zhao, D.; Song, Z.; Yuan, Z.; Li, C.; Wang, Y.; Zhou, X.; Yin, X.; Hassan, M.F.; Yang, L. HDAC6 alleviates prion peptide-mediated neuronal death via modulating PI3K-Akt-mTOR pathway. Neurobiol. Aging 2016, 37, 91–102. [Google Scholar] [CrossRef]
  228. Seo, J.-S.; Moon, M.-H.; Jeong, J.-K.; Seol, J.-W.; Lee, Y.-J.; Park, B.-H.; Park, S.-Y. SIRT1, a histone deacetylase, regulates prion protein-induced neuronal cell death. Neurobiol. Aging 2012, 33, 1110–1120. [Google Scholar] [CrossRef]
  229. Jeong, J.-K.; Moon, M.-H.; Lee, Y.-J.; Seol, J.-W.; Park, S.-Y. Autophagy induced by the class III histone deacetylase Sirt1 prevents prion peptide neurotoxicity. Neurobiol. Aging 2012, 34, 146–156. [Google Scholar] [CrossRef]
  230. Wang, J.; Zhang, J.; Shi, Q.; Zhang, B.-Y.; Chen, C.; Chen, L.-N.; Sun, J.; Wang, H.; Xiao, K.; Dong, X.-P. Scrapie Infection in Experimental Rodents and SMB-S15 Cells Decreased the Brain Endogenous Levels and Activities of Sirt1. J. Mol. Neurosci. 2014, 55, 1022–1030. [Google Scholar] [CrossRef]
  231. Boese, A.S.; Saba, R.; Campbell, K.; Majer, A.; Medina, S.; Burton, L.; Booth, T.F.; Chong, P.; Westmacott, G.; Dutta, S.M.; et al. MicroRNA abundance is altered in synaptoneurosomes during prion disease. Mol. Cell. Neurosci. 2016, 71, 13–24. [Google Scholar] [CrossRef] [PubMed]
  232. Gao, C.; Wei, J.; Zhang, B.-Y.; Shi, Q.; Chen, C.; Wang, J.; Shi, Q.; Dong, X.-P. MiRNA expression profiles in the brains of mice infected with scrapie agents 139A, ME7 and S15. Emerg. Microbes Infect. 2016, 5, 1–10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  233. Toivonen, J.M.; Sanz-Rubio, D.; López-Pérez, Ó.; Marín-Moreno, A.; Bolea, R.; Osta, R.; Badiola, J.J.; Zaragoza, P.; Espinosa, J.-C.; Torres, J.-M.; et al. MicroRNA Alterations in a Tg501 Mouse Model of Prion Disease. Biomolecules 2020, 10, 908. [Google Scholar] [CrossRef] [PubMed]
  234. Slota, J.; Medina, S.J.; Klassen, M.; Gorski, D.; Mesa, C.M.; Robertson, C.; Mitchell, G.; Coulthart, M.B.; Pritzkow, S.; Soto, C.; et al. Identification of circulating microRNA signatures as potential biomarkers in the serum of elk infected with chronic wasting disease. Sci. Rep. 2019, 9, 19705. [Google Scholar] [CrossRef]
  235. Rubio, D.S.; López-Pérez, Ó.; de Andrés Pablo, A.; Bolea, R.; Osta, R.; Badiola, J.J.; Zaragoza, P.; Martín-Burriel, I.; Toivonen, J.M. Increased circulating microRNAs miR-342-3p and miR-21-5p in natural sheep prion disease. J. Gen. Virol. 2017, 98, 305–310. [Google Scholar] [CrossRef]
  236. Pease, D.; Scheckel, C.; Schaper, E.; Eckhardt, V.; Emmenegger, M.; Xenarios, I.; Aguzzi, A. Genome-wide identification of microRNAs regulating the human prion protein. Brain Pathol. 2018, 29, 232–244. [Google Scholar] [CrossRef] [Green Version]
  237. Burak, K.; Lamoureux, L.; Boese, A.; Majer, A.; Saba, R.; Niu, Y.; Frost, K.; Booth, S.A. MicroRNA-16 targets mRNA involved in neurite extension and branching in hippocampal neurons during presymptomatic prion disease. Neurobiol. Dis. 2018, 112, 1–13. [Google Scholar] [CrossRef]
  238. Gao, C.; Shi, Q.; Wei, J.; Zhou, W.; Xiao, K.; Wang, J.; Shi, Q.; Dong, X.-P. The associations of two SNPs in miRNA-146a and one SNP in ZBTB38-RASA2 with the disease susceptibility and the clinical features of the Chinese patients of sCJD and FFI. Prion 2018, 12, 34–41. [Google Scholar] [CrossRef] [Green Version]
  239. Kang, S.-G.; Kim, C.; Aiken, J.; Yoo, H.S.; McKenzie, D. Dual MicroRNA to Cellular Prion Protein Inhibits Propagation of Pathogenic Prion Protein in Cultured Cells. Mol. Neurobiol. 2017, 55, 2384–2396. [Google Scholar] [CrossRef]
Figure 1. Comparison of differentially methylated regions (DMRs) and positions (DMPs) in prion-like diseases. Venn diagrams indicate the number of common and unique DMRs (a) and DMPs (b) in AD, PD, ALS, and HD.
Figure 1. Comparison of differentially methylated regions (DMRs) and positions (DMPs) in prion-like diseases. Venn diagrams indicate the number of common and unique DMRs (a) and DMPs (b) in AD, PD, ALS, and HD.
Ijms 23 12609 g001
Figure 2. Venn diagrams of miRNA profiles in prion-like diseases: (a) upregulated miRNAs in prion-like disease patients; (b) downregulated miRNAs in patients suffering from prion-like diseases.
Figure 2. Venn diagrams of miRNA profiles in prion-like diseases: (a) upregulated miRNAs in prion-like disease patients; (b) downregulated miRNAs in patients suffering from prion-like diseases.
Ijms 23 12609 g002
Table 4. MicroRNAs commonly upregulated and downregulated in prion and prion-like diseases. (+) = Upregulated and (−) = Downregulated.
Table 4. MicroRNAs commonly upregulated and downregulated in prion and prion-like diseases. (+) = Upregulated and (−) = Downregulated.
miRNAPrion DiseasesADPDALSHDUpregulated (+)/Downregulated (−)
miR-5100+ ++ Upregulated
miR-342-3p+/−+/−+/− Upregulated/Downregulated
let-7f-5p+ ++ Upregulated
miR-146b-5p++++ Upregulated
let-7a-5p++ + Upregulated
miR-378c++ + Upregulated
miR-27a-3p++ + Upregulated
miR-339-3p++ + Upregulated
miR-142-5p++++ Upregulated
miR-146a-5p++ Upregulated
miR-320a-3p+/−++ Upregulated/Downregulated
miR-10a-5p+ +Upregulated
miR-326+ + Upregulated
miR-21-5p+ + Upregulated
miR-324-5p+ + Upregulated
miR-103a-3p+ + Upregulated
miR-331-3p+/− +/− Upregulated/Downregulated
miR-107+ + Upregulated
miR-142-3p+ + Upregulated
miR-129-5p Downregulated
miR-423-5p Downregulated
miR-125a-5p Downregulated
miR-148a-3p Downregulated
miR-186-5p Downregulated
miR-141-3p Downregulated
miR-149-5p Downregulated
miR-200a-3p Downregulated
miR-200b Downregulated
miR-323-3p Downregulated
miR-338-3p Downregulated
miR-342-5p Downregulated
miR-382 Downregulated
miR-383 Downregulated
miR-433 Downregulated
miR-455-5p Downregulated
let-7b Downregulated
let-7c Downregulated
miR-486-3p Downregulated
miR-183-5p Downregulated
miR-100-5p Downregulated
miR-125b-5p Downregulated
miR-99a-5p Downregulated
miR-145-3p Downregulated
miR-410-3p Downregulated
miR-181d-5p Downregulated
miR-375 Downregulated
miR-99b-5p Downregulated
miR-30e-3p Downregulated
miR-129-2-3p Downregulated
miR-223-3p Downregulated
miR-493-3p Downregulated
miR-182-3p Downregulated
miR-877-5p Downregulated
miR-182-5p Downregulated
miR-144-5p Downregulated
miR-181c-5p Downregulated
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Hernaiz, A.; Toivonen, J.M.; Bolea, R.; Martín-Burriel, I. Epigenetic Changes in Prion and Prion-like Neurodegenerative Diseases: Recent Advances, Potential as Biomarkers, and Future Perspectives. Int. J. Mol. Sci. 2022, 23, 12609. https://doi.org/10.3390/ijms232012609

AMA Style

Hernaiz A, Toivonen JM, Bolea R, Martín-Burriel I. Epigenetic Changes in Prion and Prion-like Neurodegenerative Diseases: Recent Advances, Potential as Biomarkers, and Future Perspectives. International Journal of Molecular Sciences. 2022; 23(20):12609. https://doi.org/10.3390/ijms232012609

Chicago/Turabian Style

Hernaiz, Adelaida, Janne Markus Toivonen, Rosa Bolea, and Inmaculada Martín-Burriel. 2022. "Epigenetic Changes in Prion and Prion-like Neurodegenerative Diseases: Recent Advances, Potential as Biomarkers, and Future Perspectives" International Journal of Molecular Sciences 23, no. 20: 12609. https://doi.org/10.3390/ijms232012609

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