Hostname: page-component-cb9f654ff-9b74x Total loading time: 0 Render date: 2025-08-31T03:40:22.115Z Has data issue: false hasContentIssue false

MicroRNA Dysregulation in Neurodegeneration: Role, Biomarker Potential and Therapeutics

Published online by Cambridge University Press:  14 July 2025

Sunny Ramesh Dogra*
Affiliation:
Biophysical Lab, Indian Institute of Technology, Department of Chemistry, New Delhi, Delhi, India
Jyoti Bhonsle
Affiliation:
RNA Biology Lab, Indian Institute of Technology, Department of Chemistry, New Delhi, Delhi, India
Ashutosh Sharma
Affiliation:
RNA Biology Lab, Indian Institute of Technology, Department of Chemistry, New Delhi, Delhi, India
*
Corresponding author: Sunny Ramesh Dogra; Email: cyz248137@iitd.ac.in
Rights & Permissions [Opens in a new window]

Abstract

Neurodegenerative diseases (NDDs) are a group of complex disorders marked by pathophysiological mechanisms involving protein aggregation, mitochondrial dysfunction, oxidative stress and neuroinflammation. Irrespective of extensive research advances, NDDs have become a serious global concern and persist as a major therapeutic challenge. In recent years, microRNAs (miRNAs), a class of small non-coding RNAs, have established a pivotal role in combating NDDs. The altered expression of miRNAs is reported to be associated with the progression of various NDDs. This review aims to discuss miRNA biogenesis; dysregulation in NDDs, specifically Alzheimer’s disease, Parkinson’s disease (PD) and amyotrophic lateral sclerosis; their potential as biomarkers; and promising therapeutic targets. Additionally, there are various emerging technologies discussed that are advanced approaches to enhance miRNA-based diagnostics and therapeutics.

Résumé

RÉSUMÉ

La dysrégulation des micro-ARN dans la neurodégénérescence : rôle, biomarqueur potentiel et traitements. Les maladies neurodégénératives (MND) forment un groupe de troubles complexes, portant la marque de mécanismes physiopathologiques qui font intervenir l’agrégation des protéines, le dysfonctionnement mitochondrial, le stress oxydatif et la neuro-inflammation. Malgré tous les progrès de la recherche, même approfondie, les MND sont devenues un sérieux problème de santé dans le monde et continuent à poser des défis thérapeutiques importants. Au cours des dernières années, les micro ARN, classe de petits ARN non codants, se sont taillé une place de premier plan dans la lutte contre les MND. L’expression inappropriée des micro ARN serait associée à l’évolution de différentes MND. La présente revue documentaire a pour objets la biogenèse des micro ARN; leur dysrégulation dans les MND, notamment dans la maladie d’Alzheimer, la maladie de Parkinson et la sclérose latérale amyotrophique; leur potentiel de biomarqueur ainsi que les cibles thérapeutiques d’avenir. Il sera aussi question de différentes techniques de pointe permettant d’améliorer les diagnostics et les traitements fondés sur les micro ARN.

Information

Type
Review Article
Creative Commons
Creative Common License - CCCreative Common License - BYCreative Common License - SA
This is an Open Access article, distributed under the terms of the Creative Commons Attribution-ShareAlike licence (https://creativecommons.org/licenses/by-sa/4.0/), which permits re-use, distribution, and reproduction in any medium, provided the same Creative Commons licence is used to distribute the re-used or adapted article and the original article is properly cited.
Copyright
© The Author(s), 2025. Published by Cambridge University Press on behalf of Canadian Neurological Sciences Federation

Introduction

Neurodegenerative diseases (NDDs) such as Alzheimer’s disease (AD), Parkinson’s disease (PD), amyotrophic lateral sclerosis (ALS) and Huntington’s disease are characterized by progressive neuronal loss, leading to cognitive and motor impairments. Reference Dugger and Dickson1 The World Health Organization estimates that neurodegenerative disorders could surpass cancer as a leading cause of death in the coming decades. Reference Li, Lei and Sun2 Despite extensive research, effective therapeutic strategies remain limited due to the complexity of disease mechanisms. Therefore, it is important to improve our understanding of molecular mechanisms underlying neurodegeneration and identify new therapeutic targets for the prevention and treatment of NDDs. Over the past decades, miRNAs have been identified as significantly important in the context of NDDs.

MicroRNAs (miRNAs) are small, non-coding RNA molecules that have emerged as essential gene regulators at the post-transcriptional level. The evolution began in 1993 with the discovery of the first miRNA, lin-4, in Caenorhabditis elegans by Victor Ambros and Gary Ruvkun, highlighting its role in gene expression regulation. Reference Lee, Feinbaum and Ambros3,Reference Wightman, Ha and Ruvkun4 In 2001, the term “microRNA” came into recognition as a distinct class of gene regulators. Reference Lagos-Quintana, Rauhut, Lendeckel and Tuschl5,Reference Lau, Lim, Weinstein and Bartel6 The first direct evidence linking miRNAs to NDDs was demonstrated by Anne Schaefer and colleagues in 2007, who observed that the absence of Dicer, an enzyme important in miRNA biogenesis, caused progressive neurodegeneration in mouse cerebellar Purkinje cells, that is, mouse neuron cells. Reference Schaefer, O’Carroll and Tan7 Following this, in 2013, Schaefer’s team identified the key role of miRNA-128 in modulating excitability and motor behavior in mice. Reference Tan, Plotkin and Venø8 This suggested the potential role of miRNA in neurological disorders. Further, several studies indicated dysregulation of specific miRNAs in major NDDs, such as Alzheimer’s and Parkinson’s, marking them as promising biomarkers and therapeutic agents. An important breakthrough came in 2024, when Victor Ambros and Gary Ruvkun were awarded the Nobel Prize in Medicine for “the discovery of miRNA and its role in post-transcriptional gene regulation,” which emphasized the role of miRNAs in understanding and potentially treating a wide range of diseases, including NDDs. Reference Gui, Liu, Zhang, Lv and Hu9

The involvement of miRNAs in NDDs is attributed to their role in regulating crucial cellular pathways. They influence various processes, including neurogenesis, apoptosis, oxidative stress response and inflammation, all of which are key factors in various NDDs. They are abundantly present in the central nervous system, and their altered expression results in the pathogenesis of numerous NDDs. Reference Salta, Sierksma, Vanden Eynden and De Strooper10 In AD, miRNAs have been linked to the regulation of amyloid-beta (Aβ) accumulation and tau protein hyperphosphorylation, both of which are hallmarks of the disease. Reference Bekris and Leverenz11 Similarly, in PD, miRNAs affect the survival of dopaminergic neurons and mitochondrial function, further enhancing disease progression. Reference Subramaniam and Chesselet12 The dysregulation of miRNA networks contributes to impaired autophagy, lysosomal dysfunction and abnormal post-translational modifications, all of which promote pathological protein accumulation.

Beyond their role in pathogenesis, miRNAs also serve as valuable biomarkers for early diagnosis and disease monitoring and as promising therapeutic agents. Reference Azam, Rößling and Geithe13 Recent findings suggest that specific miRNA signatures can differentiate between healthy individuals and patients with NDDs, offering new avenues for precision medicine in neurology. Reference Mancuso, Hernis, Agostini, Rovaris, Caputo and Clerici14 This review aims to provide a comprehensive overview of the role of miRNAs in NDDs, highlighting their involvement in disease pathogenesis, their potential as biomarkers and the emerging therapeutic strategies targeting miRNA dysregulation.

MiRNA: an overview

MiRNAs are a class of non-coding RNAs with a size range of 20–25 nucleotides. Reference Li, Fu, Guo, Du, Chen and Cheng15 The basic structure of miRNAs begins with the transcription of primary miRNAs (pri-miRNAs) by RNA polymerase II, which are processed into precursor miRNAs (pre-miRNAs) by a complex composed of the RNase III enzyme Drosha and its binding partner DiGeorge syndrome critical region gene 8 (DGCR8). Reference Denli, Tops, Plasterk, Ketting and Hannon16 These pre-miRNAs are exported to the cytoplasm by Exportin-5 and are recognized for cleavage by Dicer, which leads to the production of miRNA duplex. Reference Bohnsack, Czaplinski and Gorlich17 This duplex is unwound by a helicase into a single strand, that is, mature miRNA, resulting in the degradation of the complementary strand. Reference Ambrus and Frolov18 Mature miRNAs then couple with the RNA-induced silencing complex, which causes post-transcriptional gene silencing by recognizing the 3’ untranslated region of the target mRNA (Figure 1). In this process, miRNAs bind to target mRNAs, preventing them from being translated into target protein and thus causing degradation of mRNAs. Reference Schwarz and Zamore19Reference Friedman, Farh, Burge and Bartel21

Figure 1. Schematic representation of miRNA biogenesis and gene silencing action of miRNA. MiRNA = microRNA; pri-miRNA = primary microRNA; pre-miRNA = precursor miRNA; DGCR8 = DiGeorge syndrome critical region gene 8; TRBP = transactivation response RNA-binding protein; RISC-RNA = induced silencing complex; mRNA = messenger RNA. Model generated with icons and templates from BioRender.com.

Though miRNAs are known for their role in inhibiting gene expression via post-transcriptional gene silencing, they also have an important role in promoting gene expression. Moreover, their role is also highlighted in the regulation of various cellular processes, either by targeting mitochondrial transcription Reference Lehmann, Krüger and Park22 or by interacting with other non-coding RNAs like long non-coding RNAs and circular RNAs. Reference Matsui, Chu and Zhang23 In addition to this, some pri-miRNAs are found to encode peptides having distinct functional roles, which are known as miRNA-encoded peptides. Reference Lauressergues, Couzigou and Clemente24,Reference Dragomir, Knutsen and Calin25

Beyond this, several studies have shown that miRNAs contribute to the pathogenesis of various neurodegenerative disorders by influencing excitotoxicity, oxidative stress, mitochondrial dysfunction, neuroinflammation and disruption of the blood-brain barrier (BBB). Reference Singh, Kukreti, Saso and Kukreti26,Reference Lin and Beal27 In general, miRNAs, such as miR-144, have been known to be upregulated in the aging brain and affect the gene expression and DNA damage response pathway, which may further lead to neurological diseases. Reference Zhou, Zhao and Zheng28 Various miRNAs, such as miR-9 and miR-146a, are involved in AD, contributing to Aβ accumulation, tau tangles and neuroinflammation, while miR-34b/c and miR-133b affect mitochondrial dysfunction in Parkinson’s disease. Reference Hébert, Horré and Nicolaï29,Reference Higgs and Lehman30

Recent research suggests that miRNAs hold promise as both biomarkers and therapeutic targets for NDDs. Their stability in biological fluids, combined with their distinct expression patterns in different neurodegenerative conditions, makes them attractive candidates for diagnostic tools. In addition to their diagnostic potential, miRNAs hold promise as therapeutic agents. Moreover, advancements in system biology and CRISPR-based technologies Reference Lehallier, Gate and Schaum31 significantly contribute to a deeper understanding of miRNA functions and regulatory mechanisms of various NDDs.

MiRNAs in various neurodegenerative diseases (NDDs)

Alzheimer’s disease

AD is a progressive neurodegenerative disorder characterized by cognitive decline and behavioral impairments. Reference Lane, Hardy and Schott32 It is a common form of dementia in the elderly and a major global concern. A survey in the USA alone states that an estimated 6.7 million elderly people currently live with Alzheimer’s dementia, a number that is expected to double by 2060 in the absence of medical advancements. 33 The main pathological hallmarks of AD are Aβ deposition and tau hyperphosphorylation, resulting in amyloid plaques and neurofibrillary tangles (NFTs), respectively. Moreover, synaptic dysfunction and neuroinflammation also contribute to disease progression. Reference Wei, Wang, Ma, Zhang, Cao and Li34 Despite advances in understanding AD, early diagnosis and effective treatments remained challenging, and an urgent need for early biomarkers carved a path for miRNAs. Various findings have illustrated the role of miRNAs in AD due to their involvement in Aβ metabolism and tau hyperphosphorylation.

The accumulation of Aβ plaques is controlled by the cleavage pathway of amyloid precursor protein (APP), a large transmembrane glycoprotein. In the non-amyloidogenic pathway, APP is sequentially cleaved by α-secretase and γ-secretase, generating neuroprotective fragments (sAPPα, p3 and amyloid intracellular domain), while in the amyloidogenic pathway, β-secretase (BACE1) and γ-secretase process APP into Aβ40 and Aβ42 (Figure 2). Aβ42 is more prone to aggregation and produces neurotoxic plaques. Reference Vu Nguyen35 Various miRNAs are responsible for the regulation of this process by varying BACE1 expression. miR-339-5p, miR-29c, miR-15b, miR-195 and miR-124 suppress BACE1 activity, leading to a reduction in Aβ production, Reference Das, Wang and Ganguly36,Reference Selkoe and Hardy37 but the downregulation of miR-339-5p in AD leads to excessive Aβ accumulation and accelerates plaque formation, Reference Long, Ray and Lahiri38 whereas miR-135b and miR-29c enhance Aβ clearance and act as neuroprotective factors. Reference Zhang, Xing, Guo, Zheng, Wang and Xu39

Figure 2. Schematic representation of amyloidogenic and non-amyloidogenic pathway of amyloid precursor protein. sAPPβ = soluble amyloid precursor protein beta; sAPPα = soluble amyloid precursor protein alpha; AICD = amyloid intracellular domain; Aβ = amyloid-beta. Model generated with icons and templates from BioRender.com.

Another important feature of AD is tau hyperphosphorylation. Tau is a microtubule-associated protein, which stabilizes the neuronal cytoskeleton, but in AD, it undergoes abnormal hyperphosphorylation by kinases like CDK5 and GSK-3β, due to which it detaches from microtubules and aggregates as NFTs. Studies have shown that miRNAs such as mir-219-5p and mir-124-3p target the kinases responsible for tau phosphorylation and regulate tau metabolism, Reference Li, Chen, Yi and Tong40,Reference Zhou, Deng, Chu, Zhao and Guo41 whereas upregulation of miR-146a promotes tau hyperphosphorylation. Reference Wang, Huang and Wang42 Moreover, dysregulation of several other miRNAs contributes to AD pathology, like mir-132, a consistently altered miRNA in AD that influences tau stability and neural apoptosis, Reference Salta, Sierksma, Vanden Eynden and De Strooper10 and mir-9, a highly conserved brain-enriched miRNA linked to tau acetylation and aggregation, as it is involved in neurofilament regulation. Reference Schonrock, Humphreys, Preiss and Götz43

Beyond these key pathological hallmarks, neuroinflammation and synaptic dysregulation are also main contributors to AD progression. Mir-146a modulates inflammatory pathways by targeting key immune regulators that amplify neuroinflammation, promoting neuronal damage, and simultaneously plays an important role in Aβ metabolism. Beyond plaque formation, the soluble Aβ oligomers enhance glutamate release, disrupting synaptic signaling and leading to excitotoxicity and neuronal death. Reference Edwards44 The neuronal function is preserved by miRNAs like mir-431 and mir-188-5p, which protect synapses by regulating Wnt/β-catenin signaling. Reference Lee, Kim and An45

There are several miRNAs that are promising biomarkers for early diagnosis. Mir-125b, mir-146a and mir-34a are consistently altered in AD patients, while mir-9, mir-118 and mir-1256 show increased expression in AD brains, suggesting their potential in identifying disease progression (Table 1). A comprehensive meta-analysis by Yoon et al. (2022) further emphasized the diagnostic value of miRNAs in AD. Reference Yoon, Kim and Ko46 It covered a wide range of samples, including blood and CSF and identified 56 miRNAs – 40 upregulated and 16 downregulated with significant differential expression in Alzheimer’s patients compared to healthy. Moreover, a study by Swarbrick et al. (2019) identified 10 miRNAs that are consistently dysregulated in AD. Reference Swarbrick, Wragg, Ghosh and Stolzing47 These miRNAs were reported to show altered expression up to 20 years before symptoms appear, which highlighted their potential as early biomarkers. Other than diagnostics, miRNA-based therapies, including miRNA mimics (which restore neuroprotective miRNA levels) and miRNA oligonucleotides (which inhibit pathogenic miRNAs), offer a novel strategy for AD treatment. Reference Bouchie48,Reference Rupaimoole and Slack49

Table 1. Dysregulation of various miRNAs in different NDDs

AD = Alzheimer’s disease; PD = Parkinson’s disease; ALS = amyloid lateral sclerosis; BACE1 = beta-site amyloid precursor protein ceaving enzyme 1; APP = amyloid precursor protein; ERK1/2 = extracellular signal-regulated kinase 1 and 2; MFN2 = mitofusin 2; ER = endoplasmic reticulum; BAG2 = Bcl-2-associated athanogene 2; GSK-3β = glycogen synthase kinase 3 beta; CDK5 = cyclin-dependent kinase 5; HSPB8 = heat shock protein family B member 8; NF-κB = nuclear factor kappa-light-chain-enhancer of activated B cells; PINK1 = PTEN-induced putative kinase 1; PBMCs = peripheral blood mononuclear cells; ELAVL4 = embryonic lethal abnormal vision-like protein 4; COX IV = cytochrome c oxidase subunit IV; PGC-1 α = peroxisome proliferator-activated receptor gamma coactivator 1 α; HDAC4 = histone deacetylase 4; Bcl-2 = B-cell lymphoma 2; Mcl-1 = myeloid cell leukemia 1; Snc = substantia nigra pars compacta; SNCA = synuclein alpha; LRRK2 = leucine-rich repeat kinase 2; SOCS1 = suppressor of cytokine signaling 1; FADD = Fas-associated protein with death domain; IKK = IkB kinase; MMP14 = matrix metalloproteinase 14; LRRN3 = leucine-rich repeat neuronal protein.

Amyotrophic lateral sclerosis

ALS, also known as Lou Gehrig’s disease, is a fatal NDD that primarily affects the upper and lower motor neurons in the brain and spinal cord. Upper and lower motor neurons are key components of the motor system, controlling voluntary and involuntary muscle movements. Reference Hardiman, Al-Chalabi and Chio50 Any damage to these neurons can lead to muscle weakness, atrophy, respiratory failure and eventual death within 2–5 years after diagnosis. ALS predominantly affects individuals aged 40–60 years. The annual incidence is approximately 2.6 cases per 100,000 people, with males exhibiting a higher risk with 1:350 compared to females with 1:400. Reference Wijesekera and Leigh51,Reference Al-Chalabi and Hardiman52 There are two types of ALS: familial ALS (accounts for approximately 10% ALS cases), which has a family history and sporadic ALS without any family history (accounts for approximately 90% ALS cases). Reference R., A. and J.53 Clinically, symptoms of familial and sporadic ALS are very similar, which include dysarthria, dysphagia, laryngeal dysfunction, abnormal neurons, pseudobulbar effects, abnormal muscles, etc. Reference Abe, Itoyama and Sobue54

The etiology of ALS is not yet well understood. Several studies indicate that the disease is associated with the aggregation of pathological proteins such as superoxide dismutase 1 (SOD1), TAR DNA-binding protein 43 (TARDBP) and fused in sarcoma (FUS). Reference F., J., Cooper-Knock, J. and Kirby55 Additionally, defining features of ALS include neuronal intracytoplasmic inclusion bodies and a hexanucleotide repeat expansion in the C9ORF72 gene located on chromosome 9. Reference Al-Chalabi, Jones, Troakes, King, Al-Sarraj and van den Berg56Reference Tomar, Sharma, Siddhanta and Deep58 In addition to these genetic factors, several hypothetical mechanisms may contribute to the disease’s progression (Figure 3). These include defects in nucleocytoplasmic transport, altered RNA metabolism, protein aggregation, abnormal DNA repair processes, dysfunctional mitochondria, issues with axonal transport, irregular vesicle transport, neuron inflammation, excitotoxicity and dysfunctional oligodendrocytes. Reference Morgan and Orrell59Reference Mejzini, Flynn, Pitout, Fletcher, Wilton and Akkari61

Figure 3. Factors leading to motor neuron degeneration in amyotrophic lateral sclerosis (ALS) patients. FUS = fused in sarcoma; SOD1 = superoxide dismutase 1; C9orf72 = chromosome 9 open reading frame 72; TDP43 = TAR DNA-binding protein 43. Model generated with icons and templates from BioRender.com.

Recent advances in RNA biology have highlighted the role of miRNAs in the pathogenesis of ALS. Research has revealed a link between the dysfunctional activity of miRNAs and ALS-related proteins. ALS-related proteins like FUS and TDP-43 have been found to influence miRNA regulation, while miRNAs, in turn, regulate both ALS-related proteins and associated genes. Reference Ricci, Marzocchi and Battistini62,Reference Freischmidt, Müller, Ludolph and Weishaupt63 Several studies have shown that specific miRNAs are altered in patients with ALS (Table 1), suggesting that these small ncRNAs may influence ALS pathology.

Parisi et al. (2016) highlighted the role of upregulated miR-125b in enhancing NF-kB signaling in microglia, promoting neuroinflammation and motor neuron damage in ALS patients. Although miR-155 upregulation impairs microglial function, miRNAs like miR-335-5p, miR-27b-3p and miR-34a-5p impair mitochondrial function by disrupting mitophagy, leading to oxidative stress and neuronal death. Reference Parisi, Napoli and Amadio64Reference Nguyen, Kumar, Fedele, Bonanno and Bonifacino67 MiR-129-5p disrupts neuronal mRNA regulation by downregulating ELAVL4, impairing motor neuron function. Reference Bronicki and Jasmin68,Reference Loffreda, Nizzardo and Arosio69 MiR-206’s overexpression may reflect a compensatory response to muscle degeneration by promoting neuromuscular regeneration, potentially delaying disease progression. Reference Przanowska, Sobierajska and Su70 Additionally, miR-181 promotes apoptosis and neuroinflammation by inhibiting mitophagy and targeting anti-apoptotic proteins. Reference Ouyang, Lu, Yue and Giffard71

Several studies have identified potential miRNA biomarkers for the early diagnosis of ALS. Liu et al. (2022) highlighted dysregulated miRNAs including miR-181c-5p, miR-27b-3p, miR-194, miR-b2122, miR-126-5p, miR-425-5p, miR-146b, miR-124a, miR-18b-5p and miR-155. Reference Liu, Zhou and Guan72 Similarly, Ruffo et al. (2023) observed significant downregulation of miR-132-5p, miR-132-3p and miR-143-3p and upregulation of miR-143-5p and miR-574-5p in sporadic ALS patients or those with mutations like TARDBP, FUS and C9ORF72. Reference Ruffo, Catalano, La Bella and Conforti73 For individuals with sporadic ALS or specific gene mutations such as TARDBP, FUS and C9ORF72, the levels of miR-132-5p and miR-132-3p (two forms of miR-132) are significantly lower. However, in cases where the patient has a mutation in the SOD1 gene, the levels of miR-132 remain unchanged. Reference Freischmidt, Müller, Ludolph and Weishaupt63 Additionally, the presence of certain miRNAs might act as early biomarkers for ALS, such as miR-132-5p, miR-132-3p, miR-124-3p and miR-133a-3p in the blood act as early biomarkers for ALS in people with a specific gene mutation (G376D-TARDBP). Reference Ruffo, Catalano, La Bella and Conforti73 Moreover, systematic analysis by Liu et al. (2023) speculated the increased expression levels of miR-206, miR113b and miR338-p as potential promising biomarkers for ALS, Reference Liu, Lan and Shi74 whereas Casado Gama et al. (2024) have identified consistently dysregulated miRNAs in various biological fluids of ALS patients. Reference Casado Gama, Amorós and Andrade de Araújo75 These include hsa-miR-3665, hsa-miR-4530, hsa-miR-4745-5p and hsa-miR-206 in serum; hsa-miR-338-3p and hsa-miR-183-5p in blood; hsa-miR-34a-3p in CSF; hsa-miR-206 in plasma; and hsa-miR-146a-5p, hsa-miR-151a-5p, hsa-miR-10b-5p, hsa-miR-29b-3p and hsa-miR-4454 in neural-enriched extracellular vesicles from plasma. The upregulation of hsa-miR-206, hsa-miR-338-3p, hsa-miR-146a-5p and hsa-miR-151a-5p and the downregulation of hsa-miR-183-5p, hsa-miR-10b-5p, hsa-miR-29b-3p and hsa-miR-4454 are reliable biomarkers for ALS across independent studies.

Parkinson’s disease

Parkinson’s disease (PD) is one of the most common neurodegenerative disorders, affecting more than 6 million people around the globe, caused by loss of dopaminergic neurons (Figure 4) in the substantia nigra pars compacta (SNc), a part of the midbrain that plays a crucial role in movement regulation, dopamine production and supporting the brain’s reinforcement network. Reference Dorsey and Bloem76,Reference Lees, Hardy and Revesz77 Degeneration of these dopaminergic neurons in the SNc can lead to characteristic symptoms of PD such as tremors, muscle inflexibility and impaired motor function. Reference Kalia and Lang78,Reference Parkinson79

Figure 4. Schematic representation of dopamine level at synaptic cleft. The level is considerably higher in (a) healthy person compared to (b) Parkinson patient.

The ailment is defined by the intracellular accumulation of α-synuclein (α-syn), manifested as Lewy bodies and Lewy neurites. Reference Jahan, Ahmad and Deep80,Reference Xu and Pu81 The precise etiology of PD remains unidentified; however, age is considered the most substantial risk factor. It is influenced by both environmental and genetic variables. Reference Pang, Ho and Liu82 α-synuclein (SNCA) and leucine-rich repeat kinase 2 (LRRK2) are the genes linked to PD; for early onset, these genes are Parkin (PARK2), oncogene DJ1 (DJ1) and PTEN-induced putative kinase 1 (PINK1). Reference Coppedè83 The pathophysiology of PD has been shown to be closely related to the dysregulation of these genes and their corresponding proteins, which are regulated by various miRNAs that ultimately promote α-synuclein accumulation, mitochondrial dysfunction, oxidative stress, calcium homeostasis, iron accumulation and neuroinflammation. Reference Poewe, Seppi and Tanner84

Recent studies have identified specific miRNA signatures that correlate with PD progression (Table 1). For example, the upregulation of miR-7 and miR-153 can repress SNCA expression, which leads to decrease in α-synuclein aggregation. Reference McMillan, Murray and Bengoa-Vergniory85Reference Wu, Xu and Zhou88 In contrast, the downregulation of miR-34b and miR-34c also suppresses DJ-1/Parkin, impairs the ubiquitin-proteasome system and reduces clearance of damaged proteins, leading to mitochondrial dysfunction and neuronal apoptosis. Additionally, the upregulation of hsa-miR-4639 5p can also inhibit DJ-1, promoting cell death. Reference Kabaria, Choi, Chaudhuri, Mouradian and Junn89Reference Rezaei, Nateghinia, Estiar, Taheri and Ghafouri-Fard91 MiR-205 supresses LRRK2, a kinase associated with both familial and sporadic PD that influences mitochondrial dynamics when mutated. It is found that miR-205 is downregulated in PD patients. Reference Mortiboys, Johansen, Aasly and Bandmann92Reference Cho, Liu and Jin94 Pro-inflammatory cytokines are released by activated microglia due the upregulation of mir-155, which causes neuroinflammation. Thome et al. (2016) highlighted that the absence of miR-155 reduces the release of α-Syn, thereby alleviating microgliosis in a sequential manner. Reference Kim, Jou and Joe95Reference Thome, Harms, Volpicelli-Daley and Standaert97 Upregulation of miR-30e reduces neuroinflammation. Reference Li, Yang, Ma, Luo, Chen and Gu98,Reference Miñones-Moyano, Porta and Escaramís90 Oxidative stress plays a central role in dopaminergic neuron death in PD. Reference Sanders and Timothy Greenamyre99 MiR-7, miR-9 and miR-29b protect against oxidative stress by targeting genes, which regulate stress responses such as Nrf2 and Sirtuin (silent mating type information regulation 2 homolog), Reference Hou, Zuo, Li, Zhang and Teng100,Reference Kabaria, Choi, Chaudhuri, Jain, Li and Junn101 whereas miR-124 enhances autophagy by targeting Bim. Reference Wang, Ye and Zhu102 Additionally, miR-181a influences autophagy by regulating the PTEN/AKT pathway. miR-29c-3p, miR-19b-3p and miR-29a-3p show decreased expression in serum from PD patients, with levels correlating with disease severity. Reference Bai, Tang and Yu103,Reference Fernández-Santiago, Iranzo and Gaig104 MiR-24 and miR-485-5p are differentially expressed in the CSF of PD patients compared to controls. Reference Marques, Kuiperij and Bruinsma105Reference Wang, Li, Wang and Su107 Notably, miR-200a-3p levels in CSF could distinguish PD patients from healthy controls with high sensitivity and specificity. A combined panel of upregulated miRNAs (miR-1275, miR-23a-5p, miR-432-5p, miR-4433b-3p, miR-4443) and downregulated miRNAs (miR-142-5p, miR-143-3p, miR-374a-3p, miR-542-3p, miR-99a-5p) achieved remarkable diagnostic accuracy. Reference Salemi, Marchese and Lanza108

Systematic studies like Santos-Lobato et al. (2021) identified approximately 99 differentially expressed miRNAs in brain samples from patients with PD, comprising 60 upregulated and 39 downregulated miRNAs, which regulate around 135 target genes; notably, hsa-miR-144 is the sole miRNA exhibiting both upregulation and downregulation in PD. Reference Santos-Lobato, Vidal and Ribeiro-dos-Santos109 Salemi et al. (2022) identified a dysregulation of the miRNAs hsa-miR-1275, hsa-miR-23a-5p, hsa-miR-432-5p, hsa-miR-4433b-3p, hsa-miR-4443, hsa-miR-142-5p, hsa-miR-143-3p, hsa-miR-374a-3p, hsa-miR-542-3p and hsa-miR-99a-5p for the first time in a study involving patients with Parkinson’s disease. Reference Salemi, Marchese and Lanza108

The alteration of miRNAs presents intriguing therapeutic methods for PD. For instance, miR-146a, miR-124 and miR-21 can inhibit inflammatory processes, miR-195 inhibits the release of pro-inflammatory cytokines and enhances the release of anti-inflammatory cytokines and miR-let-7a mitigates α-synuclein-induced microglial inflammation. Reference Slota and Booth96Reference Dorval and Hébert111 MiR-150 functions as a diagnostic biomarker by inhibiting AKT3, hence diminishing neuroinflammation and apoptosis, Reference Li, Yu and Li112 whereas miR-7, miR-7116-5p, miR-29c/30e and miR-425 also offer promising therapeutic targets for PD. Reference Li, Bi, Han and Huang113Reference Cao, Wang, Qu, Kang and Yang117 These miRNAs serve a dual purpose as biomarkers for early diagnosis and as therapeutic agents to influence critical pathways in Parkinson’s disease, such as neuroinflammation, mitochondrial failure and protein aggregation.

Therapeutic potential and biomarker applications of miRNAs

MiRNAs are small RNA molecules that help control gene expression. Over the past decade, they have become important not only in understanding how diseases work but also in developing new ways to diagnose and treat them. In complex diseases like Alzheimer’s and Parkinson’s, miRNAs offer a new approach because they can affect many genes at once, unlike traditional drugs that usually target just one protein.

Therapeutic use of miRNAs

Scientists are currently exploring two main ways to use miRNAs for therapy:

  1. 1. Inhibiting harmful miRNAs that are found at high levels in disease, using tools like antagomirs or sponges.

  2. 2. Restoring helpful miRNAs that are reduced in disease by giving synthetic copies called miRNA mimics.

For example, miR-124, which is mostly found in the brain, supports neuron health and reduces inflammation. In diseases like Alzheimer’s, levels of miR-124 drop. Studies in mice have shown that bringing back miR-124 using mimics can protect brain cells and improve memory. Reference An, Gong, Wang, Bian, Yu and Wei143 Another miRNA, miR-155, increases during brain inflammation and can worsen damage. Blocking miR-155 has been shown to lower immune cell activity in the brain and help protect neurons. Reference Cardoso, Guedes, Pereira de Almeida and Pedroso de Lima144

However, a major challenge is delivering these miRNAs to the right cells in the brain. To solve this, researchers are testing viral vectors, like adeno-associated viruses, and nanoparticles, which can carry miRNAs across the BBB. Reference Saraiva, Praça, Ferreira, Santos, Ferreira and Bernardino145

miRNAs as biomarkers

One of the most promising roles of miRNAs is in diagnosis. Since they are stable in blood, CSF and saliva, they can be used as noninvasive markers to detect brain diseases early or track their progress. For instance, in AD, higher levels of miR-125b and lower levels of miR-29a have been found in patient blood samples. These changes are linked to how severe the disease is. Reference Tan, Yu and Tan146 In Parkinson’s disease, miR-153 and miR-223 are often reduced, and this may reflect nerve cell damage and brain inflammation. Reference Cardo, Coto and Mena147 Instead of using just one miRNA, scientists often combine several into a panel to improve accuracy. Some teams have even used machine learning to analyze miRNA patterns along with clinical data, which helps doctors make better predictions. Reference Parveen, Mustafa, Yadav and Kumar148,Reference Ling, Aldoghachi and Chong149

Emerging technologies: systems biology, AI and CRISPR

New technologies are changing the way we study miRNAs and use them in medicine. Three major tools are helping in this area: systems biology, artificial intelligence (AI) and CRISPR gene editing.

Systems biology

Rather than focusing on one gene at a time, systems biology looks at how everything is connected. It helps researchers see the big picture – how miRNAs fit into complex networks of genes, proteins and cell pathways. Using this method, scientists have linked changes in miRNA levels to problems like inflammation, energy failure in cells and loss of communication between neurons in diseases like Alzheimer’s. Reference Roth, Hecker and Fava150

Artificial intelligence

There’s so much biological data today that it’s hard to analyze manually. That’s where AI and machine learning come in. These tools can scan thousands of miRNA profiles and predict which ones are linked to disease. AI-based tools like miRNet and deepTarget help identify new miRNA targets and improve diagnostic tools. Reference Panwar, Omenn and Guan151

CRISPR/Cas gene editing

CRISPR, a gene-editing tool, has opened new doors in miRNA research. A version called CRISPR/Cas13 can edit RNA directly, including miRNAs, without changing the DNA. This allows scientists to silence or boost specific miRNAs safely in the lab. For example, researchers removed miR-132 in mice and found it led to memory loss and poor brain function, suggesting this miRNA is important for learning and memory. Reference Hansen, Karelina, Sakamoto, Wayman, Impey and Obrietan152Reference Zhang and Bian154

Challenges and future perspectives

The study of miRNAs in NDDs faces several challenges that impede progress. One of the primary difficulties is the complex and multifaceted regulation of miRNAs. These small RNA molecules regulate multiple target genes, which complicates the identification of their specific roles in diseases like Alzheimer’s and Parkinson’s. The redundancy and pleiotropic nature of miRNA regulation further add to this complexity, as individual miRNAs can affect various biological pathways, often with opposing effects depending on the context. Reference Juźwik, Drake and Zhang155 Additionally, the tissue-specific expression of miRNAs poses challenges. miRNAs in the brain are localized to specific regions, and accessing relevant tissues for research is often difficult due to limited availability of postmortem samples and the inability to capture dynamic changes in living patients. Reference Kos, Puppala and Cruz156,Reference Moreau, Bruse, David-Rus, Buyske and Brzustowicz157 Furthermore, the delivery of miRNA-based therapies is hindered by theBBB, a major obstacle in translating these therapies into clinical practice. While novel delivery systems, such as nanoparticles and viral vectors, are showing promise, effective and targeted delivery to the brain remains a significant challenge. Reference Gareev, Beylerli and Tamrazov158 Another concern is the potential for off-target effects, as miRNAs often regulate multiple genes, and their manipulation could unintentionally disrupt normal cellular processes. Reference Seok, Lee, Jang and Chi159 Finally, the lack of standardized protocols for miRNA extraction and analysis contributes to inconsistencies across studies, limiting the reproducibility of results and hindering the development of universal guidelines for miRNA research. Reference Moldovan, Batte, Trgovcich, Wisler, Marsh and Piper160,Reference Rice, Roberts and Burton161

Despite these challenges, the future of miRNA research in NDDs holds great promise. One exciting possibility is the use of miRNAs as noninvasive biomarkers for early diagnosis and disease progression. miRNAs are stable in body fluids like blood and CSF, making them ideal candidates for noninvasive diagnostic tools. Reference Azam, Rößling and Geithe13,Reference Noor Eddin, Hamsho and Adi162 Moreover, targeted miRNA therapies represent a potential breakthrough in treatment strategies. By either inhibiting disease-promoting miRNAs or mimicking protective ones, researchers are exploring ways to slow or even reverse the progression of NDDs like Alzheimer’s and Parkinson’s. Reference Li, Fu, Guo, Du, Chen and Cheng15,Reference Walgrave, Zhou, De Strooper and Salta163 Advances in delivery technologies are improving the feasibility of miRNA-based treatments by developing systems capable of crossing the BBB and reaching targeted brain regions. Reference Lukiw164,Reference Fu, Chen and Huang165 Personalized medicine also presents an exciting future direction, where profiling miRNAs in patients could lead to tailored treatments based on the specific molecular mechanisms underlying each individual disease. Furthermore, combining miRNA research with other molecular approaches, such as genomics and proteomics, will provide a more comprehensive understanding of NDDs and reveal new therapeutic targets.

Acknowledgments

The authors acknowledge the Council of Scientific and Industrial Research, Government of India and the Indian Council of Medical Research for the fellowship.

Author contributions

S.R.D. and J.B. contributed equally to this work. S.R.D., J.B. writing – original draft; S.R.D., J.B. and A.S. review and editing.

Funding statement

This study was funded by the Council of Scientific and Industrial Research, Government of India (grant no. 09/0086(19988)/2024-EMR-I, no.09/0086(19971)/2024-EMR-I).

Competing interests

The authors declare no competing financial interest.

Footnotes

Sunny Ramesh Dogra and Jyoti Bhonsle are equal and first authors.

References

Dugger, BN, Dickson, DW. Pathology of neurodegenerative diseases. Cold Spring Harb Perspect Biol. 2017;9(7):a028035. doi: 10.1101/cshperspect.a028035.Google Scholar
Li, S, Lei, Z, Sun, T. The role of microRNAs in neurodegenerative diseases: a review. Cell Biol Toxicol. 2023;39(1):5383. doi: 10.1007/s10565-022-09761-x.Google Scholar
Lee, RC, Feinbaum, RL, Ambros, V. The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell. 1993;75(5):843854. doi: 10.1016/0092-8674(93)90529-y.Google Scholar
Wightman, B, Ha, I, Ruvkun, G. Posttranscriptional regulation of the heterochronic gene lin-14 by lin-4 mediates temporal pattern formation in C. elegans. Cell. 1993;75(5):855862. doi: 10.1016/0092-8674(93)90530-4.Google Scholar
Lagos-Quintana, M, Rauhut, R, Lendeckel, W, Tuschl, T. Identification of novel genes coding for small expressed RNAs. Science. 2001;294(5543):853858. doi: 10.1126/science.1064921.Google Scholar
Lau, NC, Lim, LP, Weinstein, EG, Bartel, DP. An abundant class of tiny RNAs with probable regulatory roles in caenorhabditis elegans. Science. 2001;294(5543):858862. doi: 10.1126/science.1065062.Google Scholar
Schaefer, A, O’Carroll, D, Tan, CL, et al. Cerebellar neurodegeneration in the absence of microRNAs. J Exp Med. 2007;204(7):15531558. doi: 10.1084/jem.20070823.Google Scholar
Tan, CL, Plotkin, JL, Venø, MT, et al. MicroRNA-128 governs neuronal excitability and motor behavior in mice. Science. 2013;342(6163):12541258. doi: 10.1126/science.1244193.Google Scholar
Gui, Y, Liu, H, Zhang, L, Lv, W, Hu, X. Altered MicroRNA Profiles in Cerebrospinal Fluid Exosome in Parkinson Disease and Alzheimer Disease. Oncotarget. 2015;6: 3704337053.Google Scholar
Salta, E, Sierksma, A, Vanden Eynden, E, De Strooper, B. miR-132 loss de-represses ITPKB and aggravates amyloid and TAU pathology in Alzheimer’s brain. EMBO Mol Med. 2016;8(9):10051018. doi: 10.15252/emmm.201606520.Google Scholar
Bekris, LM, Leverenz, JB. The biomarker and therapeutic potential of miRNA in Alzheimer’s disease. Neurodegener Dis Manag. 2015;5(1):6174. doi: 10.2217/nmt.14.52.Google Scholar
Subramaniam, SR, Chesselet, MF. Mitochondrial dysfunction and oxidative stress in Parkinson’s disease. Prog Neurobiol. 2013;106-107:1732. doi: 10.1016/j.pneurobio.2013.04.004.Google Scholar
Azam, HMH, Rößling, RI, Geithe, C, et al. MicroRNA biomarkers as next-generation diagnostic tools for neurodegenerative diseases: a comprehensive review. Front Mol Neurosci. 2024;17:1386735. doi: 10.3389/fnmol.2024.1386735.Google Scholar
Mancuso, R, Hernis, A, Agostini, S, Rovaris, M, Caputo, D, Clerici, M. MicroRNA-572 expression in multiple sclerosis patients with different patterns of clinical progression. J Transl Med. 2015;13(1):148. doi: 10.1186/s12967-015-0504-2.Google Scholar
Li, YB, Fu, Q, Guo, M, Du, Y, Chen, Y, Cheng, Y. MicroRNAs: pioneering regulators in Alzheimer’s disease pathogenesis, diagnosis, and therapy. Transl Psychiatry. 2024;14(1):367. doi: 10.1038/s41398-024-03075-8.Google Scholar
Denli, AM, Tops, BBJ, Plasterk, RHA, Ketting, RF, Hannon, GJ. Processing of primary microRNAs by the microprocessor complex. Nature. 2004;432(7014):231235. doi: 10.1038/nature03049.Google Scholar
Bohnsack, MT, Czaplinski, K, Gorlich, D. Exportin 5 is a ranGTP-dependent dsRNA-binding protein that mediates nuclear export of pre-miRNAs. RNA. 2004;10(2):185191. doi: 10.1261/rna.5167604.Google Scholar
Ambrus, AM, Frolov, MV. The diverse roles of RNA helicases in RNAi. Cell Cycle. 2009;8(21):35003505. doi: 10.4161/cc.8.21.9887.Google Scholar
Schwarz, DS, Zamore, PD. Why do miRNAs live in the miRNP? Genes Dev. 2002;16(9):10251031. doi: 10.1101/gad.992502.Google Scholar
Ipsaro, JJ, Joshua-Tor, L. From guide to target: molecular insights into eukaryotic RNA-interference machinery. Nat Struct Mol Biol. 2015;22(1):2028. doi: 10.1038/nsmb.2931.Google Scholar
Friedman, RC, Farh, KKH, Burge, CB, Bartel, DP. Most mammalian mRNAs are conserved targets of microRNAs. Genome Res. 2009;19(1):92105. doi: 10.1101/gr.082701.108.Google Scholar
Lehmann, SM, Krüger, C, Park, B, et al. An unconventional role for miRNA: let-7 activates toll-like receptor 7 and causes neurodegeneration. Nat Neurosci. 2012;15(6):827835. doi: 10.1038/nn.3113.Google Scholar
Matsui, M, Chu, Y, Zhang, H, et al. Promoter RNA links transcriptional regulation of inflammatory pathway genes. Nucleic Acids Res. 2013;41(22):1008610109. doi: 10.1093/nar/gkt777.Google Scholar
Lauressergues, D, Couzigou, JM, Clemente, HS, et al. Primary transcripts of microRNAs encode regulatory peptides. Nature. 2015;520(7545):9093. doi: 10.1038/nature14346.Google Scholar
Dragomir, MP, Knutsen, E, Calin, GA. SnapShot: unconventional miRNA functions. Cell. 2018;174(4):10381038.e1. doi: 10.1016/j.cell.2018.07.040.Google Scholar
Singh, A, Kukreti, R, Saso, L, Kukreti, S. Oxidative stress: a key modulator in neurodegenerative diseases. Molecules. 2019;24(8):1583. doi: 10.3390/molecules24081583.Google Scholar
Lin, MT, Beal, MF. Mitochondrial dysfunction and oxidative stress in neurodegenerative diseases. Nature. 2006;443(7113):787795. doi: 10.1038/nature05292.Google Scholar
Zhou, C, Zhao, L, Zheng, J, et al. MicroRNA-144 modulates oxidative stress tolerance in SH-SY5Y cells by regulating nuclear factor erythroid 2-related factor 2-glutathione axis. Neurosci Lett. 2017;655:2127. doi: 10.1016/j.neulet.2017.06.045.Google Scholar
Hébert, SS, Horré, K, Nicolaï, L, et al. MicroRNA regulation of Alzheimer’s amyloid precursor protein expression. Neurobiol Dis. 2009;33(3):422428. doi: 10.1016/j.nbd.2008.11.009.Google Scholar
Higgs, PG, Lehman, N. The RNA world: molecular cooperation at the origins of life. Nat Rev Genet. 2015;16(1):717. doi: 10.1038/nrg3841.Google Scholar
Lehallier, B, Gate, D, Schaum, N, et al. Undulating changes in human plasma proteome profiles across the lifespan. Nat Med. 2019;25(12):18431850. doi: 10.1038/s41591-019-0673-2.Google Scholar
Lane, CA, Hardy, J, Schott, JM. Alzheimer’s disease. Eur J Neurol. 2018;25(1):5970. doi: 10.1111/ene.13439.Google Scholar
2023 Alzheimer’s disease facts and figures. Alzheimer’s & Dementia. 2023;19(4):15981695. doi: 10.1002/alz.13016.Google Scholar
Wei, W, Wang, ZY, Ma, LN, Zhang, TT, Cao, Y, Li, H. MicroRNAs in Alzheimer’s disease: function and potential applications as diagnostic biomarkers. Front Mol Neurosci. 2020;13:160. doi: 10.3389/fnmol.2020.00160.Google Scholar
Vu Nguyen, K. β-amyloid precursor protein (APP) and the human diseases. AIMS Neurosci. 2019;6(4):273281. doi: 10.3934/Neuroscience.2019.4.273.Google Scholar
Das, U, Wang, L, Ganguly, A, et al. Visualizing APP and BACE-1 approximation in neurons yields insight into the amyloidogenic pathway. Nat Neurosci. 2016;19(1):5564. doi: 10.1038/nn.4188.Google Scholar
Selkoe, DJ, Hardy, J. The amyloid hypothesis of Alzheimer’s disease at 25 years. EMBO Mol Med. 2016;8(6):595608. doi: 10.15252/emmm.201606210.Google Scholar
Long, JM, Ray, B, Lahiri, DK. MicroRNA-339-5p down-regulates protein expression of β-site amyloid precursor protein-cleaving enzyme 1 (BACE1) in human primary brain cultures and is reduced in brain tissue specimens of Alzheimer disease subjects. J Biol Chem. 2014;289(8):51845198. doi: 10.1074/jbc.M113.518241.Google Scholar
Zhang, Y, Xing, H, Guo, S, Zheng, Z, Wang, H, Xu, D. MicroRNA-135b has a neuroprotective role via targeting of β-site APP-cleaving enzyme 1. Exp Ther Med. 2016;12(2):809814. doi: 10.3892/etm.2016.3366.Google Scholar
Li, J, Chen, W, Yi, Y, Tong, Q. miR-219-5p inhibits tau phosphorylation by targeting TTBK1 and GSK-3β in Alzheimer’s disease. J Cell Biochem. 2019;120(6):99369946. doi: 10.1002/jcb.28276.Google Scholar
Zhou, Y, Deng, J, Chu, X, Zhao, Y, Guo, Y. Role of post-transcriptional control of Calpain by miR-124-3p in the development of Alzheimer’s disease. J Alzheimers Dis. 2019;67(2):571581. doi: 10.3233/JAD-181053.Google Scholar
Wang, G, Huang, Y, Wang, LL, et al. MicroRNA-146a suppresses ROCK1 allowing hyperphosphorylation of tau in Alzheimer’s disease. Sci Rep. 2016;6(1):26697. doi: 10.1038/srep26697.Google Scholar
Schonrock, N, Humphreys, DT, Preiss, T, Götz, J. Target gene repression mediated by miRNAs miR-181c and miR-9 both of which are down-regulated by amyloid-β. J Mol Neurosci. 2012;46(2):324335. doi: 10.1007/s12031-011-9587-2.Google Scholar
Edwards, FA. A unifying hypothesis for Alzheimer’s disease: from plaques to neurodegeneration. Trends Neurosci. 2019;42(5):310322. doi: 10.1016/j.tins.2019.03.003.Google Scholar
Lee, K, Kim, H, An, K, et al. Replenishment of microRNA-188-5p restores the synaptic and cognitive deficits in 5XFAD mouse model of Alzheimer’s disease. Sci Rep. 2016;6(1):34433. doi: 10.1038/srep34433.Google Scholar
Yoon, S, Kim, SE, Ko, Y, et al. Differential expression of microRNAs in Alzheimer’s disease: a systematic review and meta-analysis. Mol Psychiatry. 2022;27(5):24052413. doi: 10.1038/s41380-022-01476-z.Google Scholar
Swarbrick, S, Wragg, N, Ghosh, S, Stolzing, A. Systematic review of miRNA as biomarkers in Alzheimer’s disease. Mol Neurobiol. 2019;56(9):61566167. doi: 10.1007/s12035-019-1500-y.Google Scholar
Bouchie, A. First microRNA mimic enters clinic. Nat Biotechnol. 2013;31(7):577577. doi: 10.1038/nbt0713-577.Google Scholar
Rupaimoole, R, Slack, FJ. MicroRNA therapeutics: towards a new era for the management of cancer and other diseases. Nat Rev Drug Discov. 2017;16(3):203222. doi: 10.1038/nrd.2016.246.Google Scholar
Hardiman, O, Al-Chalabi, A, Chio, A, et al. Amyotrophic lateral sclerosis. Nat Rev Dis Primers. 2017;3:17071. doi: 10.1038/nrdp.2017.71.Google Scholar
Wijesekera, LC, Leigh, PN. Amyotrophic lateral sclerosis. Orphanet J Rare Dis. 2009;4:3. doi: 10.1186/1750-1172-4-3.Google Scholar
Al-Chalabi, A, Hardiman, O. The epidemiology of ALS: a conspiracy of genes, environment and time. Nat Rev Neurol. 2013;9(11):617628. doi: 10.1038/nrneurol.2013.203.Google Scholar
R., H, A., H, J., AO, et al. Folding and Aggregation of Cu, Zn-Superoxide Dismutase. In: Amyotrophic Lateral Sclerosis. InTech; 2012; 95:1293012933. doi: 10.5772/31629.Google Scholar
Abe, K, Itoyama, Y, Sobue, G, et al. Confirmatory double-blind, parallel-group, placebo-controlled study of efficacy and safety of edaravone (MCI-186) in amyotrophic lateral sclerosis patients. Amyotroph Lateral Scler Frontotemporal Degener. 2014;15(7-8):610617. doi: 10.3109/21678421.2014.959024.Google Scholar
F., E, J., J, Cooper-Knock, J, J., P, Kirby, J. Genetics of Familial Amyotrophic Lateral Sclerosis. In: Amyotrophic Lateral Sclerosis. InTech; 2012: 517536. doi: 10.5772/32498.Google Scholar
Al-Chalabi, A, Jones, A, Troakes, C, King, A, Al-Sarraj, S, van den Berg, LH. The genetics and neuropathology of amyotrophic lateral sclerosis. Acta Neuropathol. 2012;124(3):339352. doi: 10.1007/s00401-012-1022-4.Google Scholar
Mackenzie, IRA, Bigio, EH, Ince, PG, et al. Pathological TDP-43 distinguishes sporadic amyotrophic lateral sclerosis from amyotrophic lateral sclerosis with SOD1 mutations. Ann Neurol. 2007;61(5):427434. doi: 10.1002/ana.21147.Google Scholar
Tomar, VR, Sharma, S, Siddhanta, S, Deep, S. Biophysical and spectroscopical insights into structural modulation of species in the aggregation pathway of superoxide dismutase 1. Commun Chem. 2025;8(1):22. doi: 10.1038/s42004-025-01421-5.Google Scholar
Morgan, S, Orrell, RW. Pathogenesis of amyotrophic lateral sclerosis. Br Med Bull. 2016;119(1):8798. doi: 10.1093/bmb/ldw026.Google Scholar
Bonafede, R, Mariotti, R. ALS pathogenesis and therapeutic approaches: the role of mesenchymal stem cells and extracellular vesicles. Front Cell Neurosci. 2017;11:80. doi: 10.3389/fncel.2017.00080.Google Scholar
Mejzini, R, Flynn, LL, Pitout, IL, Fletcher, S, Wilton, SD, Akkari, PA. ALS genetics, mechanisms, and therapeutics: where are we now? Front Neurosci. 2019;13:1310. doi: 10.3389/fnins.2019.01310.Google Scholar
Ricci, C, Marzocchi, C, Battistini, S. MicroRNAs as biomarkers in amyotrophic lateral sclerosis. Cells. 2018;7(11): 219. doi: 10.3390/cells7110219.Google Scholar
Freischmidt, A, Müller, K, Ludolph, AC, Weishaupt, JH. Systemic dysregulation of TDP-43 binding microRNAs in amyotrophic lateral sclerosis. Acta Neuropathol Commun. 2013;1(1):42. doi: 10.1186/2051-5960-1-42.Google Scholar
Parisi, C, Napoli, G, Amadio, S, et al. MicroRNA-125b regulates microglia activation and motor neuron death in ALS. Cell Death Differ. 2016;23(3):531541. doi: 10.1038/cdd.2015.153.Google Scholar
De Luna, N, Turon-Sans, J, Cortes-Vicente, E, et al. Downregulation of miR-335-5P in amyotrophic lateral sclerosis can contribute to neuronal mitochondrial dysfunction and apoptosis. Sci Rep. 2020;10(1):4308. doi: 10.1038/s41598-020-61246-1.Google Scholar
Jhanji, R, Behl, T, Sehgal, A, Bungau, S. Mitochondrial dysfunction and traffic jams in amyotrophic lateral sclerosis. Mitochondrion. 2021;58:102110. doi: 10.1016/j.mito.2021.02.008.Google Scholar
Nguyen, TPN, Kumar, M, Fedele, E, Bonanno, G, Bonifacino, T. MicroRNA Alteration, Application as biomarkers, and therapeutic approaches in neurodegenerative diseases. Int J Mol Sci. 2022;23(9):4718. doi: 10.3390/ijms23094718.Google Scholar
Bronicki, LM, Jasmin, BJ. Emerging complexity of the huD/ELAVl4 gene; implications for neuronal development, function, and dysfunction. RNA. 2013;19(8):10191037. doi: 10.1261/rna.039164.113.Google Scholar
Loffreda, A, Nizzardo, M, Arosio, A, et al. miR-129-5p: a key factor and therapeutic target in amyotrophic lateral sclerosis. Prog Neurobiol. 2020;190:101803. doi: 10.1016/j.pneurobio.2020.101803.Google Scholar
Przanowska, RK, Sobierajska, E, Su, Z, et al. miR-206 family is important for mitochondrial and muscle function, but not essential for myogenesis in vitro. The FASEB Journal. 2020;34(6):76877702. doi: 10.1096/fj.201902855RR.Google Scholar
Ouyang, YB, Lu, Y, Yue, S, Giffard, RG. miR-181 targets multiple bcl-2 family members and influences apoptosis and mitochondrial function in astrocytes. Mitochondrion. 2012;12(2):213219. doi: 10.1016/j.mito.2011.09.001.Google Scholar
Liu, J, Zhou, F, Guan, Y, et al. The biogenesis of miRNAs and their role in the development of amyotrophic lateral sclerosis. Cells. 2022;11(3):572. doi: 10.3390/cells11030572.Google Scholar
Ruffo, P, Catalano, S, La Bella, V, Conforti, FL. Deregulation of plasma microRNA expression in a TARDBP-ALS family. Biomolecules. 2023;13(4):706. doi: 10.3390/biom13040706.Google Scholar
Liu, H, Lan, S, Shi, XJ, et al. Systematic review and meta-analysis on microRNAs in amyotrophic lateral sclerosis. Brain Res Bull. 2023;194:8289. doi: 10.1016/j.brainresbull.2023.01.005.Google Scholar
Casado Gama, H, Amorós, MA, Andrade de Araújo, M, et al. Systematic review and meta-analysis of dysregulated microRNAs derived from liquid biopsies as biomarkers for amyotrophic lateral sclerosis. Noncoding RNA Res. 2024;9(2):523535. doi: 10.1016/j.ncrna.2024.02.006.Google Scholar
Dorsey, ER, Bloem, BR. The Parkinson pandemic-A call to action. JAMA Neurol. 2018;75(1):910. doi: 10.1001/jamaneurol.2017.3299.Google Scholar
Lees, AJ, Hardy, J, Revesz, T. Parkinson’s disease. The Lancet. 2009;373(9680):20552066. doi: 10.1016/S0140-6736(09)60492-X.Google Scholar
Kalia, LV, Lang, AE. Parkinson’s disease. The Lancet. 2015;386(9996):896912. doi: 10.1016/S0140-6736(14)61393-3.Google Scholar
Parkinson, J. An Essay on the Shaking Palsy. J Neuropsychiatry Clin Neurosci. 2002;14(2):223236. doi: 10.1176/jnp.14.2.223,Google Scholar
Jahan, I, Ahmad, A, Deep, S. Effect of flavonoids on the destabilization of α-synuclein fibrils and their conversion to amorphous aggregate: a molecular dynamics simulation and experimental study. Biochimica et Biophysica Acta (BBA) - Proteins and Proteomics. 2023;1871(6):140951. doi: 10.1016/j.bbapap.2023.140951.Google Scholar
Xu, L, Pu, J. Alpha-synuclein in Parkinson’s disease: from pathogenetic dysfunction to potential clinical application. Parkinsons Dis. 2016;2016:110. doi: 10.1155/2016/1720621.Google Scholar
Pang, SYY, Ho, PWL, Liu, HF, et al. The interplay of aging, genetics and environmental factors in the pathogenesis of Parkinson’s disease. Transl Neurodegener. 2019;8:23. doi: 10.1186/s40035-019-0165-9.Google Scholar
Coppedè, F. Genetics and epigenetics of Parkinson’s disease. Sci World J. 2012;2012:112. doi: 10.1100/2012/489830.Google Scholar
Poewe, W, Seppi, K, Tanner, CM, et al. Parkinson disease. Nat Rev Dis Primers. 2017;3:17013. doi: 10.1038/nrdp.2017.13.Google Scholar
McMillan, KJ, Murray, TK, Bengoa-Vergniory, N, et al. Loss of microRNA-7 regulation leads to α-synuclein accumulation and dopaminergic neuronal loss in vivo. Mol Ther. 2017;25(10):24042414. doi: 10.1016/j.ymthe.2017.08.017.Google Scholar
Junn, E, Lee, KW, Jeong, BS, Chan, TW, Im, JY, Mouradian, MM. Repression of alpha-synuclein expression and toxicity by microRNA-7. Proc Natl Acad Sci U S A. 2009;106(31):1305213057. doi: 10.1073/pnas.0906277106.Google Scholar
Je, G, Kim, YS. Mitochondrial ROS-mediated post-transcriptional regulation of α-synuclein through miR-7 and miR-153. Neurosci Lett. 2017;661:132136. doi: 10.1016/j.neulet.2017.09.065.Google Scholar
Wu, L, Xu, Q, Zhou, M, et al. Plasma miR-153 and miR-223 levels as potential biomarkers in Parkinson’s disease. Front Neurosci. 2022;16:865139. doi: 10.3389/fnins.2022.865139.Google Scholar
Kabaria, S, Choi, DC, Chaudhuri, AD, Mouradian, MM, Junn, E. Inhibition of miR-34b and miR-34c enhances α-synuclein expression in Parkinson’s disease. FEBS Lett. 2015;589(3):319325. doi: 10.1016/j.febslet.2014.12.014.Google Scholar
Miñones-Moyano, E, Porta, S, Escaramís, G, et al. MicroRNA profiling of Parkinson’s disease brains identifies early downregulation of miR-34b/c which modulate mitochondrial function. Hum Mol Genet. 2011;20(15):30673078. doi: 10.1093/hmg/ddr210.Google Scholar
Rezaei, O, Nateghinia, S, Estiar, MA, Taheri, M, Ghafouri-Fard, S. Assessment of the role of non-coding RNAs in the pathophysiology of Parkinson’s disease. Eur J Pharmacol. 2021;896:173914. doi: 10.1016/j.ejphar.2021.173914.Google Scholar
Mortiboys, H, Johansen, KK, Aasly, JO, Bandmann, O. Mitochondrial impairment in patients with Parkinson disease with the G2019S mutation in LRRK2 . Neurology. 2010;75(22):20172020. doi: 10.1212/WNL.0b013e3181ff9685.Google Scholar
Wang, X, Yan, MH, Fujioka, H, et al. LRRK2 regulates mitochondrial dynamics and function through direct interaction with DLP1. Hum Mol Genet. 2012;21(9):19311944. doi: 10.1093/hmg/dds003.Google Scholar
Cho, HJ, Liu, G, Jin, SM, et al. MicroRNA-205 regulates the expression of Parkinson’s disease-related leucine-rich repeat kinase 2 protein. Hum Mol Genet. 2013;22(3):608620. doi: 10.1093/hmg/dds470.Google Scholar
Kim, JH, Jou, I, Joe, EH. Suppression of miR-155 expression in IFN-γ-treated astrocytes and microglia by DJ-1: a possible mechanism for maintaining SOCS1 expression. Exp Neurobiol. 2014;23(2):148154. doi: 10.5607/en.2014.23.2.148.Google Scholar
Slota, JA, Booth, SA. MicroRNAs in neuroinflammation: implications in disease pathogenesis, biomarker discovery and therapeutic applications. Noncoding RNA. 2019;5(2):35. doi: 10.3390/ncrna5020035.Google Scholar
Thome, AD, Harms, AS, Volpicelli-Daley, LA, Standaert, DG. microRNA-155 regulates alpha-synuclein-induced inflammatory responses in models of Parkinson disease. J Neurosci. 2016;36(8):23832390. doi: 10.1523/JNEUROSCI.3900-15.2016.Google Scholar
Li, D, Yang, H, Ma, J, Luo, S, Chen, S, Gu, Q. MicroRNA-30e regulates neuroinflammation in MPTP model of Parkinson’s disease by targeting Nlrp3. Hum Cell. 2018;31(2):106115. doi: 10.1007/s13577-017-0187-5.Google Scholar
Sanders, LH, Timothy Greenamyre, J. Oxidative damage to macromolecules in human Parkinson disease and the rotenone model. Free Radic Biol Med. 2013;62:111120. doi: 10.1016/j.freeradbiomed.2013.01.003.Google Scholar
Hou, M, Zuo, X, Li, C, Zhang, Y, Teng, Y. Mir-29b regulates oxidative stress by targeting SIRT1 in ovarian cancer cells. Cell Physiol Biochem. 2017;43(5):17671776. doi: 10.1159/000484063.Google Scholar
Kabaria, S, Choi, DC, Chaudhuri, AD, Jain, MR, Li, H, Junn, E. MicroRNA-7 activates Nrf2 pathway by targeting Keap1 expression. Free Radic Biol Med. 2015;89:548556. doi: 10.1016/J.FREERADBIOMED.2015.09.010.Google Scholar
Wang, H, Ye, Y, Zhu, Z, et al. MiR-124 regulates apoptosis and autophagy process in MPTP model of Parkinson’s disease by targeting to bim. Brain Pathol. 2016;26(2):167176. doi: 10.1111/bpa.12267.Google Scholar
Bai, X, Tang, Y, Yu, M, et al. Downregulation of blood serum microRNA 29 family in patients with Parkinson’s diseaseSci Rep. 2017;7(1):5411. doi: 10.1038/s41598-017-03887-3 Google Scholar
Fernández-Santiago, R, Iranzo, A, Gaig, C, et al. MicroRNA association with synucleinopathy conversion in rapid eye movement behavior disorder. Ann Neurol. 2015;77(5):895901. doi: 10.1002/ana.24384.Google Scholar
Marques, TM, Kuiperij, HB, Bruinsma, IB, et al. MicroRNAs in cerebrospinal fluid as potential biomarkers for Parkinson’s disease and multiple system atrophy. Mol Neurobiol. 2017;54(10):77367745. doi: 10.1007/s12035-016-0253-0.Google Scholar
Cardo, LF, Coto, E, Ribacoba, R, et al. MiRNA profile in the substantia nigra of Parkinson’s disease and healthy subjects. J Mol Neurosci. 2014;54(4):830836. doi: 10.1007/s12031-014-0428-y.Google Scholar
Wang, J, Li, HY, Wang, HS, Su, ZB. MicroRNA-485 modulates the TGF-β/ smads signaling pathway in chronic asthmatic mice by targeting Smurf2. Cell Physiol Biochem. 2018;51(2):692710. doi: 10.1159/000495327.Google Scholar
Salemi, M, Marchese, G, Lanza, G, et al. Role and dysregulation of miRNA in patients with Parkinson’s disease. Int J Mol Sci. 2022;24(1):712. doi: 10.3390/ijms24010712.Google Scholar
Santos-Lobato, BL, Vidal, AF, Ribeiro-dos-Santos, Â. Regulatory miRNA-mRNA networks in Parkinson’s disease. Cells. 2021;10(6):1410. doi: 10.3390/cells10061410.Google Scholar
Zhang, J, Zhou, Dongwei, Zhang, Z, et al. miR-let-7a suppresses α-synuclein-induced microglia inflammation through targeting STAT3 in Parkinson’s disease. Biochem Biophys Res Commun. 2019;519(4):740746. doi: 10.1016/j.bbrc.2019.08.140.Google Scholar
Dorval, V, Hébert, SS. LRRK2 in transcription and translation regulation: relevance for Parkinson’s disease. Front Neurol. 2012;3:12. doi: 10.3389/fneur.2012.00012.Google Scholar
Li, H, Yu, L, Li, M, et al. MicroRNA-150 serves as a diagnostic biomarker and is involved in the inflammatory pathogenesis of Parkinson’s disease. Mol Genet Genomic Med. 2020;8(4):e1189. doi: 10.1002/mgg3.1189.Google Scholar
Li, S, Bi, G, Han, S, Huang, R. MicroRNAs play a role in Parkinson’s disease by regulating microglia function: from pathogenetic involvement to therapeutic potential. Front Mol Neurosci. 2021;14:744942. doi: 10.3389/fnmol.2021.744942.Google Scholar
He, Q, Wang, Q, Yuan, C, Wang, Y. Downregulation of miR-7116-5p in microglia by MPP+ sensitizes TNF-α production to induce dopaminergic neuron damage. Glia. 2017;65(8):12511263. doi: 10.1002/glia.23153.Google Scholar
Hu, YB, Zhang, YF, Wang, H, et al. miR-425 deficiency promotes necroptosis and dopaminergic neurodegeneration in Parkinson’s disease. Cell Death Dis. 2019;10(8):589. doi: 10.1038/s41419-019-1809-5.Google Scholar
Zhou, Y, Lu, M, Du, RH, et al. MicroRNA-7 targets nod-like receptor protein 3 inflammasome to modulate neuroinflammation in the pathogenesis of Parkinson’s disease. Mol Neurodegener. 2016;11:28. doi: 10.1186/s13024-016-0094-3.Google Scholar
Cao, B, Wang, T, Qu, Q, Kang, T, Yang, Q. Long noncoding RNA SNHG1 promotes neuroinflammation in Parkinson’s disease via regulating miR-7/NLRP3 pathway. Neuroscience. 2018;388:118127. doi: 10.1016/j.neuroscience.2018.07.019.Google Scholar
Hébert, SS, Horré, K, Nicolaï, L, et al. Loss of microRNA cluster miR-29a/b-1 in sporadic Alzheimer’s disease correlates with increased BACE1/beta-secretase expression. Proc Natl Acad Sci U S A. 2008;105(17):64156420. doi: 10.1073/pnas.0710263105.Google Scholar
Jiao, Y, Kong, L, Yao, Y, et al. Osthole decreases beta amyloid levels through up-regulation of miR-107 in Alzheimer’s disease. Neuropharmacology. 2016;108:332344. doi: 10.1016/j.neuropharm.2016.04.046.Google Scholar
Moncini, S, Lunghi, M, Valmadre, A, et al. The miR-15/107 family of microRNA genes regulates CDK5R1/p35 with implications for Alzheimer’s disease pathogenesis. Mol Neurobiol. 2017;54(6):43294342. doi: 10.1007/s12035-016-0002-4.Google Scholar
Zhu, QB, Unmehopa, U, Bossers, K, et al. MicroRNA-132 and early growth response-1 in nucleus basalis of meynert during the course of Alzheimer’s disease. Brain. 2016;139(3):908921. doi: 10.1093/brain/awv383.Google Scholar
Banzhaf-Strathmann, J, Benito, E, May, S, et al. Micro <scp>RNA</scp> -125b induces tau hyperphosphorylation and cognitive deficits in Alzheimer’s disease. EMBO J. 2014;33(15):16671680. doi: 10.15252/embj.201387576.RNA+-125b+induces+tau+hyperphosphorylation+and+cognitive+deficits+in+Alzheimer’s+disease.+EMBO+J.+2014;33(15):1667–1680.+doi:+10.15252/embj.201387576.>Google Scholar
Millan, MJ. Linking deregulation of non-coding RNA to the core pathophysiology of Alzheimer’s disease: An integrative review. Prog Neurobiol. 2017;156:168. doi: 10.1016/j.pneurobio.2017.03.004.Google Scholar
Higaki, S, Muramatsu, M, Matsuda, A, et al. Defensive effect of microRNA-200b/c against amyloid-beta peptide-induced toxicity in Alzheimer’s disease models. PLoS One. 2018;13(5):e0196929. doi: 10.1371/journal.pone.0196929.Google Scholar
Ko, CY, Chu, YY, Narumiya, S, et al. CCAAT/enhancer-binding protein delta/miR135a/thrombospondin 1 axis mediates PGE2-induced angiogenesis in Alzheimer’s disease. Neurobiol Aging. 2015;36(3):13561368. doi: 10.1016/j.neurobiolaging.2014.11.020.Google Scholar
Remenyi, J, van den Bosch, MWM, Palygin, O, et al. miR-132/212 knockout mice reveal roles for these miRNAs in regulating cortical synaptic transmission and plasticity. PLoS One. 2013;8(4):e62509. doi: 10.1371/journal.pone.0062509.Google Scholar
Yuan, J, Wu, Y, Li, L, Liu, C. MicroRNA-425-5p promotes tau phosphorylation and cell apoptosis in Alzheimer’s disease by targeting heat shock protein B8. J Neural Transm (Vienna). 2020;127(3):339346. doi: 10.1007/s00702-019-02134-5.Google Scholar
Nagaraj, S, Want, A, Laskowska-Kaszub, K, et al. Candidate Alzheimer’s disease biomarker miR-483-5p lowers TAU phosphorylation by direct ERK1/2 repression. Int J Mol Sci. 2021;22(7):3653. doi: 10.3390/ijms22073653.Google Scholar
Butovsky, O, Jedrychowski, MP, Cialic, R, et al. Targeting mi <scp>R</scp> -155 restores abnormal microglia and attenuates disease in <scp>SOD</scp> 1 mice. Ann Neurol. 2015;77(1):7599. doi: 10.1002/ana.24304.R+-155+restores+abnormal+microglia+and+attenuates+disease+in+SOD+1+mice.+Ann+Neurol.+2015;77(1):75–99.+doi:+10.1002/ana.24304.>Google Scholar
Kim, J, Fiesel, FC, Belmonte, KC, et al. miR-27a and miR-27b regulate autophagic clearance of damaged mitochondria by targeting PTEN-induced putative kinase 1 (PINK1). Mol Neurodegener. 2016;11(1):55. doi: 10.1186/s13024-016-0121-4.Google Scholar
Tai, Y, Pu, M, Yuan, L, et al. miR-34a-5p regulates PINK1-mediated mitophagy via multiple modes. Life Sci. 2021;276:119415. doi: 10.1016/j.lfs.2021.119415.Google Scholar
Aschrafi, A, Schwechter, AD, Mameza, MG, Natera-Naranjo, O, Gioio, AE, Kaplan, BB. MicroRNA-338 regulates local cytochrome c oxidase IV mRNA levels and oxidative phosphorylation in the axons of sympathetic neurons. J Neurosci. 2008;28(47):1258112590. doi: 10.1523/JNEUROSCI.3338-08.2008.Google Scholar
Russell, AP, Wada, S, Vergani, L, et al. Disruption of skeletal muscle mitochondrial network genes and miRNAs in amyotrophic lateral sclerosis. Neurobiol Dis. 2013;49:107117. doi: 10.1016/j.nbd.2012.08.015.Google Scholar
Liu, J, Zuo, X, Han, J, et al. MiR-9-5p inhibits mitochondrial damage and oxidative stress in AD cell models by targeting GSK-3β. Biosci Biotechnol Biochem. 2020;84(11):22732280. doi: 10.1080/09168451.2020.1797469.Google Scholar
Cheng, M, Liu, L, Lao, Y, et al. MicroRNA-181a suppresses parkin-mediated mitophagy and sensitizes neuroblastoma cells to mitochondrial uncoupler-induced apoptosis. Oncotarget. 2016;7(27):4227442287. doi: 10.18632/oncotarget.9786.Google Scholar
Alizadeh, M, Safarzadeh, A, Beyranvand, F, et al. The potential role of miR-29 in health and cancer diagnosis, prognosis, and therapy. J Cell Physiol. 2019;234(11):1928019297. doi: 10.1002/jcp.28607.Google Scholar
Sharma, S, Pavlasova, GM, Seda, V, et al. miR-29 modulates CD40 signaling in chronic lymphocytic leukemia by targeting TRAF4: an axis affected by BCR inhibitors. Blood. 2021;137(18):24812494. doi: 10.1182/blood.2020005627.Google Scholar
Liu, K, Liu, J, Bo, QF. MFI2-AS1 regulates the aggressive phenotypes in glioma by modulating MMP14 via a positive feedback loop. Eur Rev Med Pharmacol Sci. 2019;23(13):58845895. doi: 10.26355/eurrev_201907_18333.Google Scholar
Sangokoya, C, Doss, JF, Chi, JT. Iron-responsive miR-485-3p regulates cellular iron homeostasis by targeting ferroportin. PLoS Genet. 2013;9(4):e1003408. doi: 10.1371/journal.pgen.1003408.Google Scholar
Jiang, F, Wu, Q, Sun, S, Bi, G, Guo, L. Identification of potential diagnostic biomarkers for Parkinson’s disease. FEBS Open Bio. 2019;9(8):14601468. doi: 10.1002/2211-5463.12687.Google Scholar
Salemi, M, Lanza, G, Mogavero, MP, et al. A transcriptome analysis of mRNAs and long non-coding RNAs in patients with Parkinson’s disease. Int J Mol Sci. 2022;23(3):1535. doi: 10.3390/ijms23031535.Google Scholar
Li, X, Su, Y, Li, N, Zhang, FR, Zhang, N. Berberine attenuates MPP+-induced neuronal injury by regulating LINC00943/miR-142-5p/KPNA4/NF-κB pathway in SK-N-SH cells. Neurochem Res. 2021;46(12):32863300. doi: 10.1007/s11064-021-03431-w.Google Scholar
An, F, Gong, G, Wang, Y, Bian, M, Yu, L, Wei, C. MiR-124 acts as a target for Alzheimer’s disease by regulating BACE1. Oncotarget. 2017;8(69):114065114071. doi: 10.18632/oncotarget.23119.Google Scholar
Cardoso, AL, Guedes, JR, Pereira de Almeida, L, Pedroso de Lima, MC. miR-155 modulates microglia-mediated immune response by down-regulating SOCS-1 and promoting cytokine and nitric oxide production. Immunology. 2012;135(1):7388. doi: 10.1111/j.1365-2567.2011.03514.x.Google Scholar
Saraiva, C, Praça, C, Ferreira, R, Santos, T, Ferreira, L, Bernardino, L. Nanoparticle-mediated brain drug delivery: overcoming blood-brain barrier to treat neurodegenerative diseases. J Control Release. 2016;235:3447. doi: 10.1016/j.jconrel.2016.05.044.Google Scholar
Tan, L, Yu, JT, Tan, MS, et al. Genome-wide serum microRNA expression profiling identifies serum biomarkers for Alzheimer’s disease. J Alzheimers Dis. 2014;40(4):10171027. doi: 10.3233/JAD-132144.Google Scholar
Cardo, LF, Coto, E, Mena, L, et al. Profile of microRNAs in the plasma of Parkinson’s disease patients and healthy controls. J Neurol. 2013;260(5):14201422. doi: 10.1007/s00415-013-6900-8.Google Scholar
Parveen, A, Mustafa, SH, Yadav, P, Kumar, A. Applications of machine learning in miRNA discovery and target prediction. Curr Genomics. 2019;20(8):537544. doi: 10.2174/1389202921666200106111813.Google Scholar
Ling, L, Aldoghachi, AF, Chong, ZX, et al. Addressing the clinical feasibility of adopting circulating miRNA for breast cancer detection, monitoring and management with artificial intelligence and machine learning platforms. Int J Mol Sci. 2022;23(23):15382. doi: 10.3390/ijms232315382.Google Scholar
Roth, W, Hecker, D, Fava, E. Systems biology approaches to the study of biological networks underlying Alzheimer’s disease: role of miRNAs. Methods Mol Biol. 2016;1303:349377. doi: 10.1007/978-1-4939-2627-5_21.Google Scholar
Panwar, B, Omenn, GS, Guan, Y. miRmine: a database of human miRNA expression profiles. Bioinformatics. 2017;33(10):15541560. doi: 10.1093/bioinformatics/btx019.Google Scholar
Hansen, KF, Karelina, K, Sakamoto, K, Wayman, GA, Impey, S, Obrietan, K. miRNA-132: a dynamic regulator of cognitive capacity. Brain Struct Funct. 2013;218(3):817831. doi: 10.1007/s00429-012-0431-4.Google Scholar
Hansen, KF, Sakamoto, K, Aten, S, et al. Targeted deletion of miR-132/-212 impairs memory and alters the hippocampal transcriptome. Learn Mem. 2016;23(2):6171. doi: 10.1101/lm.039578.115.Google Scholar
Zhang, M, Bian, Z. Alzheimer’s disease and microRNA-132: a widespread pathological factor and potential therapeutic target. Front Neurosci. 2021;15:687973. doi: 10.3389/fnins.2021.687973.Google Scholar
Juźwik, CA, Drake, S, Zhang, Y, et al. MicroRNA dysregulation in neurodegenerative diseases: a systematic review. Prog Neurobiol. 2019;182:101664. doi: 10.1016/j.pneurobio.2019.101664.Google Scholar
Kos, MZ, Puppala, S, Cruz, D, et al. Blood-based miRNA biomarkers as correlates of brain-based miRNA expression. Front Mol Neurosci. 2022;15:817290. doi: 10.3389/fnmol.2022.817290.Google Scholar
Moreau, MP, Bruse, SE, David-Rus, R, Buyske, S, Brzustowicz, LM. Altered microRNA expression profiles in postmortem brain samples from individuals with schizophrenia and bipolar disorder. Biol Psychiatry. 2011;69(2):188193. doi: 10.1016/j.biopsych.2010.09.039.Google Scholar
Gareev, I, Beylerli, O, Tamrazov, R, et al. Methods of miRNA delivery and possibilities of their application in neuro-oncology. Noncoding RNA Res. 2023;8(4):661674. doi: 10.1016/j.ncrna.2023.10.002.Google Scholar
Seok, H, Lee, H, Jang, ES, Chi, SW. Evaluation and control of miRNA-like off-target repression for RNA interference. Cell Mol Life Sci. 2018;75(5):797814. doi: 10.1007/s00018-017-2656-0.Google Scholar
Moldovan, L, Batte, KE, Trgovcich, J, Wisler, J, Marsh, CB, Piper, M. Methodological challenges in utilizing miRNAs as circulating biomarkers. J Cell Mol Med. 2014;18(3):371390. doi: 10.1111/jcmm.12236.Google Scholar
Rice, J, Roberts, H, Burton, J, et al. Assay reproducibility in clinical studies of plasma miRNA. PLoS One. 2015;10(4):e0121948. doi: 10.1371/journal.pone.0121948.Google Scholar
Noor Eddin, A, Hamsho, K, Adi, G, et al. Cerebrospinal fluid microRNAs as potential biomarkers in Alzheimer’s disease. Front Aging Neurosci. 2023;15:1210191. doi: 10.3389/fnagi.2023.1210191.Google Scholar
Walgrave, H, Zhou, L, De Strooper, B, Salta, E. The promise of microRNA-based therapies in Alzheimer’s disease: challenges and perspectives. Mol Neurodegener. 2021;16(1):76. doi: 10.1186/s13024-021-00496-7.Google Scholar
Lukiw, WJ. Fission impossible: stabilized miRNA-based analogs in neurodegenerative disease. Front Neurosci. 2022;16:875957. doi: 10.3389/fnins.2022.875957.Google Scholar
Fu, Y, Chen, J, Huang, Z. Recent progress in microRNA-based delivery systems for the treatment of human disease. ExRNA. 2019;1(1):24. doi: 10.1186/s41544-019-0024-y.Google Scholar
Figure 0

Figure 1. Schematic representation of miRNA biogenesis and gene silencing action of miRNA. MiRNA = microRNA; pri-miRNA = primary microRNA; pre-miRNA = precursor miRNA; DGCR8 = DiGeorge syndrome critical region gene 8; TRBP = transactivation response RNA-binding protein; RISC-RNA = induced silencing complex; mRNA = messenger RNA. Model generated with icons and templates from BioRender.com.

Figure 1

Figure 2. Schematic representation of amyloidogenic and non-amyloidogenic pathway of amyloid precursor protein. sAPPβ = soluble amyloid precursor protein beta; sAPPα = soluble amyloid precursor protein alpha; AICD = amyloid intracellular domain; Aβ = amyloid-beta. Model generated with icons and templates from BioRender.com.

Figure 2

Table 1. Dysregulation of various miRNAs in different NDDs

Figure 3

Figure 3. Factors leading to motor neuron degeneration in amyotrophic lateral sclerosis (ALS) patients. FUS = fused in sarcoma; SOD1 = superoxide dismutase 1; C9orf72 = chromosome 9 open reading frame 72; TDP43 = TAR DNA-binding protein 43. Model generated with icons and templates from BioRender.com.

Figure 4

Figure 4. Schematic representation of dopamine level at synaptic cleft. The level is considerably higher in (a) healthy person compared to (b) Parkinson patient.