Next Article in Journal
Current Novel Targeted Therapeutic Strategies in Multiple Myeloma
Previous Article in Journal
PARG Protein Regulation Roles in Drosophila Longevity Control
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Exploring the Regulatory Landscape of Dementia: Insights from Non-Coding RNAs

1
Department of Integrated Biological Sciences, Pusan National University, Busan 46241, Republic of Korea
2
Institute of Systems Biology, Pusan National University, Busan 46241, Republic of Korea
3
Department of Biological Sciences, College of Natural Sciences, Pusan National University, Busan 46241, Republic of Korea
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(11), 6190; https://doi.org/10.3390/ijms25116190
Submission received: 26 April 2024 / Revised: 24 May 2024 / Accepted: 3 June 2024 / Published: 4 June 2024
(This article belongs to the Section Molecular Neurobiology)

Abstract

:
Dementia, a multifaceted neurological syndrome characterized by cognitive decline, poses significant challenges to daily functioning. The main causes of dementia, including Alzheimer’s disease (AD), frontotemporal dementia (FTD), Lewy body dementia (LBD), and vascular dementia (VD), have different symptoms and etiologies. Genetic regulators, specifically non-coding RNAs (ncRNAs) such as microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs), are known to play important roles in dementia pathogenesis. MiRNAs, small non-coding RNAs, regulate gene expression by binding to the 3′ untranslated regions of target messenger RNAs (mRNAs), while lncRNAs and circRNAs act as molecular sponges for miRNAs, thereby regulating gene expression. The emerging concept of competing endogenous RNA (ceRNA) interactions, involving lncRNAs and circRNAs as competitors for miRNA binding, has gained attention as potential biomarkers and therapeutic targets in dementia-related disorders. This review explores the regulatory roles of ncRNAs, particularly miRNAs, and the intricate dynamics of ceRNA interactions, providing insights into dementia pathogenesis and potential therapeutic avenues.

1. Introduction

Dementia is a complex neurological syndrome that can significantly disrupt daily life through memory loss and impaired thinking and decision-making processes [1,2]. The incidence of dementia is on the rise, particularly before the age of 65, highlighting the importance of new strategies in diagnosis and treatment [3,4,5]. There are several causes of dementia, including Alzheimer’s disease (AD), frontotemporal dementia (FTD), Lewy body dementia (LBD), and vascular dementia (VD) [6,7]. AD, the most prevalent form of dementia, is characterized by the accumulation of beta-amyloid plaques and tau tangles in the brain, which disrupt neural connectivity and cause memory loss and progressive cognitive decline [8,9]. FTD is characterized by prominent behavioral and personality changes, often associated with abnormal protein deposits in the frontal and temporal lobes [10,11]. Unlike AD, memory loss may not be an initial symptom and individuals with FTD may exhibit socially inappropriate behaviors and language difficulties [12]. LBD is characterized by the presence of Lewy bodies, abnormal protein deposits causing cognitive fluctuations, visual hallucinations, and motor symptoms resembling Parkinson’s disease (PD) [13,14]. LBD may also involve fluctuating alertness and attention, which distinguishes it from other forms of dementia [15,16]. Finally, VD results from impaired blood flow to the brain, typically caused by vascular pathology, including strokes [17,18]. Symptoms vary depending on the affected areas of the brain and may include difficulties with planning and organizing, as well as memory impairment [19]. VD is unique in its association with cardiovascular risk factors and cerebrovascular events [7]. Each subtype of dementia presents a distinct set of challenges. Accurate diagnosis and targeted interventions in the realm of dementia care require an understanding of the characteristics, symptoms, and differences between AD, FTD, LBD, and VD [20,21].
Dementia-related diseases can be caused by genetic regulators, particularly non-coding RNAs (ncRNAs) such as microRNA (miRNA), long non-coding RNA (lncRNA), and circular RNA (circRNA) [22,23]. MiRNA, a small ncRNA of about 20 nucleotides, regulates its target gene by binding complementarily to the 3′ untranslated region (UTR) of the gene [24,25]. One of the most well-known functions of miRNA is to inhibit the expression of its target gene through degradation or translation inhibition of target messenger RNA (mRNA) [26,27]. A mature miRNA is produced from a miRNA gene in several steps. First, the miRNA is transcribed from the miRNA gene to produce primary miRNA (pri-miRNA). This pri-miRNA is then processed into precursor miRNA (pre-miRNA) by the Drosha-DiGeorge Syndrome Critical Region Gene 8 (DGCR8) complex [28,29]. The pre-miRNA is transported to the cytoplasm by exportin 5, where it is cleaved by the Dicer-TAR RNA-binding Protein (TRBP) complex to form double-stranded RNA (dsRNA) [30]. The dsRNA is unwound to produce the mature single-stranded miRNA, which associates with Argonaute (AGO) proteins to form the RNA-induced silencing complex (RISC) [31,32,33]. When miRNA binds to target genes complementarily, it can be influenced by other ncRNAs with complementary sequences of miRNA [34]. These ncRNAs act as regulators of miRNA, with lncRNA and circRNA being prominent examples. LncRNA, characterized by its length exceeding 200 nucleotides, intricately modulates gene expression by acting as a molecular sponge, decoy, or guide for miRNA [35,36]. The biogenesis of lncRNA involves a series of steps for its functional maturation. LncRNA is transcribed by RNA polymerase II and subsequently undergoes several processes, including splicing to remove introns and ligate exons, as well as post-transcriptional modifications such as RNA editing and chemical modifications. Processed lncRNA exists in either the nucleus or the cytoplasm, where it is involved in various cellular functions [37,38,39]. Meanwhile, circRNA, distinguished by its covalently closed-loop structure, also contributes to the regulatory landscape as another crucial player [40]. Transcription of genes in the nucleus initiates RNA synthesis, using the DNA sequence as a template to make precursor RNA. A unique process called backsplicing is involved in the formation of circRNA, which differs from typical linear mRNA production. During backsplicing, a downstream splice donor site (5′ end) of the precursor RNA joins an upstream splice acceptor site (3′ end), making the RNA circular closed-loop structure with no 5′ or 3′ ends. This often involves skipping one or more exons, resulting in circRNAs with specific coding sequences. The size of the circRNA varies depending on which exons are included. The biogenesis of lncRNA involves a series of steps to ensure their functional maturation or exclusion [41,42,43]. CircRNA is known for its resistance to exonucleolytic degradation and function as miRNA sponge [24,44]. Therefore, these distinctive features of circRNA make them noteworthy in the field of genetic regulation.
Recent studies have highlighted the significance of competing endogenous RNA (ceRNA), a complex network of RNA–RNA interactions, as promising targets for disease biomarkers and therapeutic interventions [45,46]. In particular, ceRNA includes regulatory elements such as lncRNA and circRNA, both acting as molecular competitors for miRNA binding. Furthermore, the intricate dynamics of ceRNA interactions have recently emerged as crucial orchestrators in the complex symphony of genetic expression [47,48]. The interactions between miRNAs and ceRNAs play an essential role in shaping the pathogenesis of dementia and hold promise as diagnostic indicators and therapeutic targets (Figure 1) [49,50]. Therefore, we discuss these interactions, focusing on the complex dynamics of ceRNAs, especially miRNAs, in dementia-related diseases and provide a comprehensive overview.

2. Dementia-Related Diseases and miRNAs

Dementia-related diseases are influenced by a multitude of genes that interact with miRNAs as regulatory elements [51]. By examining the findings of numerous previous studies, this section clarifies the role of miRNAs in the development and progression of dementia. Understanding the interaction of these factors is crucial for accurate diagnosis and personalized management to identify genetic markers and potential therapeutic targets within this complex landscape [52].

2.1. Alzheimer’s Disease and miRNA

AD is thought to be caused by a combination of genetic, age-related, and environmental factors, including the accumulation of abnormal brain proteins, neuroinflammation, vascular complications, and lifestyle choices [8,53]. Recent studies have shown that changes in gene expression, regulated by miRNAs, play an important role in AD [54,55,56]. While these miRNAs are typically present within cells, they are also secreted into extracellular fluids, influencing the pathophysiology of AD. Therefore, this subsection discusses the effects of miRNAs on AD, depending on their location.

2.1.1. Differentially Expressed miRNAs in AD

It is widely recognized that miRNAs are ubiquitous in cells and influence gene expression. Accordingly, miRNAs stand out as a focal point of investigation, not only for their regulatory role in disease pathogenesis but also as promising candidates for diagnostic and therapeutic targets (Table 1).
The upregulated miRNAs in AD, including miR-20b-5p, miR-140, miR-425-5p, and miR-132, were identified to be implicated in neuronal protection by regulating cellular apoptosis and inflammatory responses [57,58,59,60]. The significance of amyloid-beta peptide (Aβ), derived from amyloid-beta precursor protein (APP) in AD pathogenesis, was highlighted by Wang et al. Their study elucidated the complex implications of miR-20b-5p in AD, noting changes in its levels in different brain regions during disease progression. In neuronal cells, elevated levels of miR-20b-5p were observed to decrease intracellular Ca2+ transients, leading to lower cell density through decreased synaptic branching, and ultimately reduce cortical thickness. Consequently, upregulation of miR-20b-5p was associated with an increased risk of AD. Paradoxically, miR-20b-5p was also found to reduce APP levels by binding to its mRNA, although the underlying mechanism remains unclear. This result highlighted the complex interplay between miR-20b-5p, APP, and the development of AD, which warrants further investigation into their interactions [57]. Another study revealed that overexpression of miR-425-5p increased tau phosphorylation and glycogen synthase kinase-3beta (GSK-3β), leading to increased cell death. These results were obtained by correlating miR-425-5p with heat shock protein B8 (HSPB8), which is known to play a role in the removal of misfolded proteins, thereby contributing to the regulation of neurodegenerative diseases. The authors suggested that miR-425-5p could be a potential therapeutic target for AD treatment [59].
A noteworthy feature of AD is characterized by many genetic factors, especially beta-amyloid plaques and tau tangles, which are associated with the Beta-Secretase 1 (BACE1) gene and specific mutations with miRNA interactions [93]. For this reason, several miRNAs, including miR-103, miR-107, miR-149, miR-9-5p, and miR-29c-3p, which decreased in AD, were observed to target BACE1 [75,78,79,82]. These downregulations were linked to enhanced neuronal protection and growth by attenuating Aβ signaling through BACE1, thus emphasizing the potential therapeutic significance of miRNAs in AD. Recent research further supported the role of miRNA dysfunction in AD pathology, especially in increased Aβ production and impaired clearance [73]. In addition, miR-31, identified as decreasing in AD patients, has been found to simultaneously reduce APP and BACE1 mRNA levels, leading to significant improvements in memory deficits and cognitive inflexibility. Overexpression of miR-31 also attenuated AD neuropathology, suggesting that miR-31 modulation of APP and BACE1 could offer a promising therapeutic approach for AD [83]. On the other hand, there is another study carried out with a genetic factor regulated by miRNAs other than BACE1. Downregulated miR-326 reduced tau phosphorylation and prevented neuronal apoptosis by binding to its target gene Vav Guanine Nucleotide Exchange Factor 1 (VAV1) in the JNK signaling pathway. This finding suggested that miR-326 has potential as a target for AD treatment, improving cognitive function and inhibiting neuronal apoptosis VAV1 [72].

2.1.2. Exosomal miRNA and G-Quadruplex Structure in AD

Exosomal miRNAs (exo-miRs) refer to miRNAs that are released into extracellular fluids after being encapsulated within exosomes, extracellular vesicles [94,95]. Exosomes released by diverse cells play a crucial role in intercellular communication by conveying miRNAs and other substances [96,97]. Exosomes are considered carriers of miRNAs and are thought to play a critical role in the understanding, diagnosis, and treatment of AD, like cellular miRNAs [98,99,100]. The diagnostic process for AD involves a comprehensive evaluation that integrates clinical, neuropsychological, imaging, and laboratory assessments to differentiate between its various subtypes [101,102,103]. Accurate diagnosis of AD is essential, and while tissue analysis is crucial, the risks involved highlight the growing need for non-invasive detection methods. Exosomes are poised to meet this demand, particularly for early diagnosis, by utilizing differentially expressed exosomal miRNAs as key diagnostic tools. For example, subjective cognitive decline (SCD), one of the earliest symptoms of AD, is not only a symptom of memory loss, but is also highly genetically influenced, highlighting the importance of exosomal miRNAs for its early detection. The researchers suggested that peripheral neuronal-derived exosomal Aβ, tau, and upregulated exo-miR-384 were key modifiers in the pathogenesis of SCD as well as AD and could be used for early diagnosis of AD [67]. In another study, researchers demonstrated that overexpression of exo-miR-22 inhibited pyroptosis by targeting gasdermin D (GSDMD), thereby enhancing memory and motor abilities in AD through the suppression of inflammation. Given the therapeutic potential of exosomes for AD, the researchers hypothesized that exo-miR-22 could have a positive impact on AD outcomes [87].
Meanwhile, there is a structural study of the relationship between miRNA and AD. The G-quadruplex, a structure found in nucleic acids with guanine-rich sequences, is known to play a crucial role in protein binding and is thought to be a target for several diseases [104,105,106]. One study showed that the AD-associated single nucleotide polymorphism (SNP), rs2291418, located within pre-miR-1229, led to the formation of a G-quadruplex structure in equilibrium with the hairpin structure. In individuals with the rs2291418 in pre-miR-1229, there was an increase in the mature form of pre-miR-1229, miR-1229-3p, in AD cases. The miR-1229-3p bound complementarily to sortilin-related receptor 1 (SORL1), which is involved in the processing and trafficking of Aβ. The study demonstrated that pre-miR-1229 has not only a typical hairpin but also a G-quadruplex secondary structure, which leads to the production of a large amount of miR-1229-3p and a decrease in the expression of SORL1. The identification of a G-quadruplex structure within pre-miR-1229 represents a promising avenue for therapeutic intervention in AD [68].

2.2. Other Dementia-Related Diseases and miRNA

Although AD is the most commonly recognized cause of dementia, there are several other types of dementia such as FTD, LBD, and VD [107]. Due to the low incidence of these dementia-related diseases excluding AD, research on the role of miRNAs is limited compared to AD [108]. Nonetheless, some studies have explored genetic factors and miRNA regulation in these other types of dementia (Table 2). The current research landscape reveals a significant gap in our understanding of the mechanisms and treatment options for FTD, LBD, and VD when compared to AD [109]. Therefore, urgent attention is required to conduct further studies and investigations into the underlying mechanisms and therapeutic strategies in FTD, LBD, and VD. Such efforts have the potential to expand our knowledge base and ultimately improve patient outcomes in these complex neurological disorders.

2.2.1. Differentially Expressed miRNAs in FTD

First of all, FTD is a neurodegenerative disorder characterized by progressive damage to the frontal and temporal lobes, with genetic factors being recognized as major contributors to its onset [127]. Several publications have focused on identifying miRNAs targeting these genetic factors in the quest for potential therapeutic interventions. Upregulated miRNAs like miR-29b and miR-659-3p were representatively target granulin precursor (GRN) which is known to be involved in FTD progression [110,111]. Jiao et al. confirmed that the absence of progranulin (PGRN), which is encoded by GRN, is implicated in certain types of FTD, a significant neurodegenerative disease with early onset. The mechanisms governing PGRN expression are not well-understood. The researchers found that ectopic expression of miR-29b reduced PGRN levels, and induced progranulin deficiency, leading to FTD. Also, they suggested that targeting miR-29b or other miRNAs could be a novel therapeutic approach to increase PGRN levels in individuals with FTD, as a post-transcriptional regulator of PGRN [110]. On the other hand, MiR-632, known to be associated with mutant GRN, but limited in functional studies, showed diagnostic potential for the disease. Significantly downregulated in the cerebrospinal fluid (CSF) of FTD patients compared to healthy controls, the decreased expression of miR-632 suggests its utility as a valuable diagnostic biomarker for both genetic and sporadic forms of FTD, distinguishing them from AD and healthy individuals [112].

2.2.2. Differentially Expressed miRNAs in LBD

LBD is characterized by the presence of abnormal protein deposits called Lewy bodies in the brain, leading to progressive cognitive decline, visual hallucinations, and motor symptoms resembling PD [128]. However, it differs from PD in several ways [129,130,131,132]. In LBD, cognitive impairment and visual hallucinations often precede or occur alongside motor symptoms, whereas PD primarily exhibits motor symptoms like tremors, stiffness, and bradykinesia, with cognitive decline emerging later in the disease progression [129]. The distinct temporal presentation of symptoms sets these two conditions apart despite their shared feature of Lewy bodies. Differences in genetic factors, such as the alpha-synuclein (SNCA) gene, also play distinct roles in PD and LBD [130,133]. Therefore, there is a need to identify key markers, such as miRNAs, to differentiate LBD from PD in diagnosis. While research on miRNAs in LBD is scarce, there is a study aimed at distinguishing LBD from PD based on miRNA expression patterns. The researchers explained that it is uncertain whether miR-7-5p directly decreases SNCA expression, but higher levels of miR-7-5p may contribute to maintaining baseline SNCA mRNA expression. Additionally, the short structural variant rs777296100-poly-T in SNCA, which may affect miRNA binding, was moderately associated with LBD but not with PD. This research also suggested that the miR-7/SNCA axis may be a potential diagnostic marker for LBD, as it is significantly upregulated in LBD compared to normal [116].

2.2.3. Differentially Expressed miRNAs in VD

Finally, VD is characterized by cognitive decline resulting from impaired blood flow to the brain, often arising as the second most common cause of dementia after AD [134]. Unlike other dementia-related diseases, VD is associated with cerebrovascular disease and vascular-related brain damage, leading to disruptions in cognitive function [135]. The key distinguishing feature of VD is its vascular etiology, where strokes or other vascular events contribute to the onset and progression of VD [136]. Therefore, understanding the underlying vascular factors is crucial for accurate diagnosis and targeted interventions in cases of VD. In this regard, research on miRNAs in VD offers promise for understanding complex genetic factors and exploring the potential for diagnosis and treatment. For example, miR-210-5p, miR-134, miR-150, and miR-181a are upregulated in VD, resulting in synaptic loss and impairing spatial learning, memory functions, speech, and language, which also contribute to associated neurodevelopmental disorders [117,118,120]. For example, one study aimed to investigate the impact of miRNAs on early VD. MiR-210-5p was increased in the hippocampus of rats exposed to 4 weeks of ischemia and binds complementarily to synaptosomal-associated protein of 25 KDa (Snap25) mRNA, causing cognitive deficits and synaptic loss, suggesting a potential new therapeutic avenue for the treatment of VD [117]. Forkhead box P2 (Foxp2), which is inhibited by miR-134-5p, is a transcription factor crucial for vocal learning. Elevated miR-134-5p levels were observed in the cortex of VD model rats, antagomir of miR-134-5p significantly mitigated synaptic protein loss by upregulating Foxp2. This implicated the miR-134-5p/Foxp2 axis in early VD-related cognitive decline [118]. Furthermore, miR-150 was investigated for the impacts on VD. The results suggested that reducing miR-150 levels may alleviate VD symptoms, significantly improving cognitive impairment and reducing neuron apoptosis in the brain by upregulating homeobox A1 (HOXA1) expression [120]. Additionally, miR-134 was found to decrease Cofilin 2 levels, thereby regulating oxidative stress and autophagy in the brain. Suppression of miR-134 in VD rats improved cognitive function by modulating oxidative stress and autophagy, resulting in induced levels of Cofilin 2 [121]. Another study revealed that downregulated miR-181a can increase PTEN-induced kinase 1 (PINK1)/Parkin expression and enhanced mitophagy to improve mitochondrial function and enhance cognitive capabilities in VD. These findings highlight the potential of miR-181a modulation to attenuate cognitive decline by regulating mitophagy processes in VD [123].
Conversely, downregulated miRNAs such as miR-132-3p, miR-322-5p, and miR-181a were associated with inflammatory responses and cognitive improvement in VD [123,125,126]. For instance, one study assessed the levels of miR-132-3p using a VD mouse model induced by bilateral carotid artery occlusion. The overexpression of miR-132-3p led to the downregulation of RAS P21 Protein Activator 1 (RASA1) expression that influenced the Ras/Akt/GSK-3β pathway, leading to improved neuronal survival and neurite outgrowth [125]. In addition, miR-322-5p was found to alleviate the development of VD by targeting tetraspanin 5 (TSPAN5), which may play a critical role in modulating cognitive function and mitophagy in the context of VD. TSPAN5, a member of the tetraspanin family, is known for its involvement in cell signaling, growth, and motility. Studies have implicated it in VD pathology, including activation of ADAM10 and the Notch pathway. In experimental models of neuronal injury and cognitive dysfunction, TSPAN5 expression was elevated and its overexpression attenuated the effects of miR-322-5p on neuronal injury. Conversely, the knockdown of TSPAN5 alleviated cognitive dysfunction in rats subjected to cerebral ischemia. This is a valuable finding that may provide a potential regulatory mechanism for studying cognitive function [126]. These downregulated miRNAs are considered potential treatments for VD, highlighting the importance of studying genetic factors of VD.

3. Dementia-Related Diseases and Other Non-Coding RNAs

The significance of ceRNA and its relevance to dementia has garnered increasing attention in the scientific community, particularly as it has been revealed that ceRNA interactions involving lncRNA or circRNA, miRNA, and mRNA play a critical role in the regulation of gene expression [137,138,139]. In the context of dementia, understanding these intricate ceRNA networks holds promise for unraveling the molecular mechanisms underlying the disease [140,141,142,143]. Investigating how these RNA molecules interact and influence each other can provide valuable insights into potential diagnostic markers and therapeutic targets for dementia [144]. The exploration of ceRNA networks may contribute to advancing our comprehension of the complex RNA-mediated regulatory pathways implicated in dementia pathogenesis.

3.1. LncRNA

LncRNAs are a diverse group of non-protein-coding transcripts that exhibit distinct features. They play a significant role in regulating gene expression through various mechanisms, including acting as sponges for miRNAs and functioning as ceRNAs [145,146,147,148]. In this capacity, lncRNAs possess miRNA response elements, allowing them to sequester and competitively inhibit miRNA activity. This role enables lncRNAs to modulate the availability of miRNAs for their target mRNAs, thereby influencing post-transcriptional gene expression regulation [149,150]. Understanding these distinctive features and the involvement of lncRNAs in miRNA sponging mechanisms contributes to unraveling complex regulatory networks. Importantly, these mechanisms have implications for various biological contexts, including their potential relevance to dementia [151]. In particular, some studies have explored the roles of lncRNAs Metastasis Associated Lung Adenocarcinoma Transcript 1 (MALAT1), also known as non-coding Nuclear-Enriched Abundant Transcript (NEAT) 2, and NEAT1 in dementia [152,153]. The role of these lncRNAs in dementia is expected to involve complex regulatory functions [154]. These findings contribute to a growing body of research exploring the functions of MALAT1, NEAT1, and various other lncRNAs in the context of dementia-related diseases (Figure 2, Table S1).
Some research has been conducted on lncRNAs, sponging miRNAs that target BACE1, a key gene involved in AD, which accounts for about 80% of dementia cases [155]. MiR-374b-5p, miR-9-5p, and miR-27a-3p were representative examples and one of the studies investigated the regulatory role between lncRNA, Membrane Associated Guanylate Kinase, WW, and PDZ Domain Containing 2 Antisense RNA 3 (MAGI2-AS3), and miR-374b-5p in Aβ-induced neurotoxicity and neuroinflammation in AD [156,157,158,159]. MiR-374b-5p targets BACE1 and MAGI2-AS3 functions as a sponge for miR-374b-5p. Upon exposure to Aβ, there was an increase in MAGI2-AS3 and a decrease in miR-374b-5p levels in neuronal and microglial cells. Reduction of MAGI2-AS3 and overexpression of miR-374b-5p alleviated neurotoxicity and neuroinflammation, while miR-374b-5p inhibition reverses these effects. Serum levels of MAGI2-AS3 and miR-374b-5p in Alzheimer’s patients were negatively correlated and associated with disease severity, suggesting the MAGI2-AS3/miR-374b-5p axis as a potential biomarker and therapeutic target for AD [156]. Another study highlights the role of Brain-Derived Neurotrophic Factor-AS (BDNF-AS) in the pathogenesis of AD by demonstrating its elevated expression in the peripheral blood of AD patients, correlating with cognitive decline. Elevated BDNF-AS promoted neurotoxicity by enhancing BACE1 expression through competitive binding with miR-9-5p, facilitating amyloid deposition and cognitive impairment in AD mice. Additionally, miR-27a-3p and NEAT1 showed consistent expression trends in AD patients, with decreased levels of miR-27a-3p and increased NEAT1 in serum correlating with disease severity and Aβ deposition, suggesting their involvement in AD progression [158].
In FTD, transactivation response DNA binding protein 43 kDa (TDP-43) is thought to be one of the causes of disease onset, along with GRN. Although TDP-43 is known to be a nuclear RNA-binding protein, its targeting and processing of mRNA are not well-understood [160]. Studies have predicted that mislocalized TDP-43 contributes to neurodegeneration in FTD by regulating the stability of mRNA such as GRN [161]. One study revealed that in the brain of FTD patients, TDP-43 was significantly increased and that TDP-43-regulated mRNAs largely control neural development or encode proteins associated with neurological disorders. Also, RNA-binding analysis results showed that TDP-43 tended to bind to the lncRNAs MALAT1 and NEAT1. Highlighting the importance of TDP-43 in regulating mRNAs through lncRNAs, understanding the interaction of TDP-43 with RNA is crucial for insights into the neurodegenerative processes of FTD [162].
VD, the second leading cause of dementia, also involves mechanisms related to lncRNA [163]. Exosomes containing the lncRNA Myocardial Infarction-Associated Transcript (MIAT) can improve cognitive function in VD by enhancing hippocampal pathology, reducing oxidative stress, and downregulating inflammatory and amyloid-beta-related markers. The therapeutic effects of MIAT were implicated with miR-34b-5p [164]. On the other hand, elevated levels of MALAT1 in VD were found to correlate with deficits in spatial learning and memory, as well as a reduction in miR-9-3p. In addition, synapse-associated protein-97 (SAP97) was found to be upregulated in the hippocampus of those with VD. In vitro experiments on hippocampal neurons showed that miR-9-3p negatively regulated SAP97 expression. Furthermore, the downregulation of MALAT1 increased miR-9-3p and reduced SAP97, whereas inhibition of miR-9-3p rescued the reduction in SAP97. This study suggests that MALAT1 upregulated SAP97 by targeting miR-9-3p in the hippocampus of VD mice, shedding light on the molecular mechanisms underlying VD [165].
While investigations into lncRNA have been conducted in various dementia-related disorders, the majority of research has been concentrated on AD. However, the available studies are limited and there is a noticeable bias towards AD. Moreover, no significant research on lncRNA in the context of LBD has been identified. lncRNAs have potential as useful diagnostic markers and therapeutic targets in diseases; further extensive research is needed to improve our understanding of these conditions [155,166,167].

3.2. CircRNA

CircRNAs are used as versatile regulators due to their unique characteristics, such as resistance to degradation, stability, and widespread presence across species [168,169,170]. They exhibit tissue-specific expression patterns and dynamic regulation during various physiological and pathological processes [171,172]. Functionally, circRNAs act as miRNA sponges, interact with RNA-binding proteins, and influence alternative splicing [168]. Several studies in AD have investigated the association between circRNAs and miRNAs, yet similar research in other dementia-related disorders is limited (Figure 3, Table S2) [171,173,174,175,176,177].
For instance, downregulation of hsa_circ_002048 was found to upregulate three miRNAs (hsa-miR-422a, hsa-miR-4784, and hsa-miR-3944-3p), potentially limiting the expression of adaptor-related protein complex 2 subunit mu 1 (AP2M1). This dysregulation led to impaired endocytosis, including autophagy, resulting in elevated proinflammatory cytokines and neurotoxic β-amyloid levels. These cascading events contributed to inflammation, β-amyloid deposition, and tau hyperphosphorylation, impacting AD progression. While offering insights into AD pathogenesis, this study suggested that these ncRNAs could serve as potential targets for diagnostic biomarkers, paving the way for advancements in AD diagnosis and treatment [178]. Another study focused on identifying dysregulated circRNA, hsa_circ_0003391, in the peripheral blood of AD patients and its association with clinical manifestations. Hsa_circ_0003391 was specifically downregulated in AD patients compared to other dementia types. The study demonstrated a significant reduction in hsa_circ_0003391 expression in the peripheral blood of AD patients, correlating with the clinical characteristics of AD. This study identified hsa_circ_0003391 as a potential biomarker for AD diagnosis and suggested insights for improving diagnostic approaches. Additionally, the research offered a new perspective for investigating the pathogenesis of AD, potentially leading to the development of innovative therapies targeting ncRNA [179]. In a specific case related to BACE1, a study aimed to investigate the influence of circ-AXL on neuronal injury and inflammation in cellular models of AD, and to elucidate the underlying molecular mechanisms. The study revealed that overexpression of circ-AXL resulted in increased apoptosis, reduced neurite outgrowth, and elevated levels of inflammatory cytokines in cellular AD models. Circ-AXL was found to negatively regulate miR-328 and positively modulate BACE1 expression. Conversely, miR-328 had a negative regulatory effect on BACE1. Overexpression of miR-328 reduced apoptosis, promoted neurite outgrowth, and decreased inflammatory cytokine levels in cellular AD models, while knockdown of miR-328 had the opposite effect. Notably, the attenuation of miR-328 reduced the impact of circ-AXL knockdown on cellular AD models. Furthermore, upregulation of BACE1 exacerbated neuronal injury and inflammation, counteracting the effects of miR-328 overexpression in cellular AD models. These findings suggested that circ-AXL may be a promising therapeutic target in AD due to its regulation of BACE1 through miR-328 [180].
Despite the unique characteristics and functional roles of circRNAs, research on their involvement in other forms of dementia, excluding AD, is limited. Therefore, there is a significant gap in understanding circRNA involvement in other dementia-related diseases. Thorough investigations into the features and functions of circRNAs in various dementia-related conditions are necessary to comprehend their role in neurodegenerative processes fully. Such research is crucial for uncovering the potential diagnostic, prognostic, and therapeutic implications of circRNAs, advancing dementia research beyond AD.

4. Dementia Pathogenesis with Genetic Factors and ncRNA Insights

The study aimed to give a comprehensive overview of the involvement of diverse genetic factors in dementia-related diseases and to explore their correlations with genes and ncRNAs. In particular, the relationship between key regulators in each dementia-related disease was visualized (Figure 4). Specifically in AD, overexpression of BACE1 leading to Aβ accumulation was regarded as a primary cause. In addition, the regulation of miRNAs targeting BACE1 and the role of lncRNAs and circRNAs as miRNA sponges were key regulatory factors in AD (Figure 4a) [53,93,157,180]. Research in FTD has primarily focused on the correlation between GRN and TDP-43, with limited simultaneous studies on their regulation by ncRNAs. Therefore, this study concentrated on predicting the correlation between mislocalized TDP-43, which regulates GRN, and related miRNAs and lncRNAs (Figure 4b) [110,111,162]. LBD is characterized by misfolded α-synuclein due to splicing errors, but there have been few studies on miRNA involvement. Therefore, this study focused on the only miRNA research about miR-7 and miR-153, which regulate exon-skipped SNCA mRNA binding and are expected to inhibit LBD (Figure 4c) [116]. Finally, VD is linked to vascular damage with various implicated genetic factors. However, research on the key regulators that mainly function in pathogenesis is limited. Therefore, we tried to envision the need for interaction to identify these key regulators concerned with ncRNAs (Figure 4d) [181].

5. Conclusions

Dementia, which includes a broad category of neurocognitive disorders characterized by progressive decline, indicates several diseases such as AD, FTD, LBD, and VD. Although numerous factors contribute to the development of dementia-related diseases, genetic factors regulated by ncRNAs play a critical role in their pathogenesis. While many studies have been conducted on how ncRNAs affect dementia in animal models, there are limitations as these models do not fully mimic human conditions. In addition, although there are some studies using brain tissue from patients, it is difficult to obtain living brain tissue, and the tissue used in research is generally severely damaged, reducing the accuracy of the research. To address this issue, recent studies have focused on ncRNAs present in serum, which is relatively easier to obtain and does not require surgical procedures. Nevertheless, there is still a lack of research on ncRNAs involved in the signaling pathways associated with the development of dementia-related diseases. Therefore, mechanistic studies on ncRNAs in dementia-related diseases are necessary. The accumulation of these studies could ultimately contribute to the development of effective diagnostic and therapeutic strategies for dementia by identifying clinically important ncRNAs and elucidating their mechanisms.

Supplementary Materials

The supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ijms25116190/s1. References [20,156,157,158,159,162,164,165,172,173,174,175,176,177,178,179,180,182,183,184,185,186,187,188] are cited in the Supplementary Materials.

Author Contributions

For Conceptualization, H.-S.K. and J.-m.K.; Writing the original draft, J.-m.K.; Review and Editing: W.R.K., E.G.P., D.H.L., Y.J.L., H.J.S., H.-s.J. and H.-Y.R. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

This work was supported by a 2-Year Research Grant of Pusan National University.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Browne, B.; Kupeli, N.; Moore, K.J.; Sampson, E.L.; Davies, N. Defining end of life in dementia: A systematic review. J. Palliat. Med. 2021, 35, 1733–1746. [Google Scholar] [CrossRef] [PubMed]
  2. Hegde, S.; Ellajosyula, R. Capacity issues and decision-making in dementia. Ann. Indian Acad. Neurol. 2016, 19 (Suppl. S1), S34–S39. [Google Scholar] [PubMed]
  3. Liang, C.S.; Li, D.J.; Yang, F.C.; Tseng, P.T.; Carvalho, A.F.; Stubbs, B.; Thompson, T.; Mueller, C.; Shin, J.I.; Radua, J.; et al. Mortality rates in Alzheimer’s disease and non-Alzheimer’s dementias: A systematic review and meta-analysis. Lancet Healthy Longev. 2021, 2, e479–e488. [Google Scholar] [CrossRef] [PubMed]
  4. Prince, M.; Ali, G.C.; Guerchet, M.; Prina, A.M.; Albanese, E.; Wu, Y.T. Recent global trends in the prevalence and incidence of dementia, and survival with dementia. Alzheimer’s Res. Ther. 2016, 8, 23. [Google Scholar] [CrossRef] [PubMed]
  5. Carter, J. Prevalence of all cause young onset dementia and time lived with dementia: Analysis of primary care health records. J. Dement. Care 2022, 30, 1–5. [Google Scholar]
  6. Chaudhry, A.; Houlden, H.; Rizig, M. Novel fluid biomarkers to differentiate frontotemporal dementia and dementia with Lewy bodies from Alzheimer’s disease: A systematic review. J. Neurol. Sci. 2020, 415, 116886. [Google Scholar] [CrossRef] [PubMed]
  7. Tayler, H.; Miners, J.S.; Güzel, Ö.; MacLachlan, R.; Love, S. Mediators of cerebral hypoperfusion and blood-brain barrier leakiness in Alzheimer’s disease, vascular dementia and mixed dementia. Brain Pathol. 2021, 31, e12935. [Google Scholar] [CrossRef] [PubMed]
  8. Lei, P.; Ayton, S.; Bush, A.I. The essential elements of Alzheimer’s disease. J. Biol. Chem. 2021, 296, 100105. [Google Scholar] [CrossRef] [PubMed]
  9. Rajmohan, R.; Reddy, P.H. Amyloid-beta and phosphorylated tau accumulations cause abnormalities at synapses of Alzheimer’s disease neurons. J. Alzheimer’s Dis. 2017, 57, 975–999. [Google Scholar] [CrossRef] [PubMed]
  10. Antonioni, A.; Raho, E.M.; Lopriore, P.; Pace, A.P.; Latino, R.R.; Assogna, M.; Mancuso, M.; Gragnaniello, D.; Granieri, E.; Pugliatti, M.; et al. Frontotemporal Dementia, Where Do We Stand? A Narrative Review. Int. J. Mol. Sci. 2023, 24, 11732. [Google Scholar] [CrossRef] [PubMed]
  11. Pressman, P.S.; Miller, B.L. Diagnosis and management of behavioral variant frontotemporal dementia. Biol. Psychiatry 2014, 75, 574–581. [Google Scholar] [CrossRef] [PubMed]
  12. Musa, G.; Slachevsky, A.; Muñoz-Neira, C.; Méndez-Orellana, C.; Villagra, R.; González-Billault, C.; Ibáñez, A.; Hornberger, M.; Lillo, P. Alzheimer’s Disease or Behavioral Variant Frontotemporal Dementia? Review of Key Points Toward an Accurate Clinical and Neuropsychological Diagnosis. J. Alzheimer’s Dis. 2020, 73, 833–848. [Google Scholar] [CrossRef] [PubMed]
  13. Utsumi, K.; Fukatsu, R.; Yamada, R.; Takamaru, Y.; Hara, Y.; Yasumura, S. Characteristics of initial symptoms and symptoms at diagnosis in probable dementia with Lewy body disease: Incidence of symptoms and gender differences. Psychogeriatrics 2020, 20, 737–745. [Google Scholar] [CrossRef] [PubMed]
  14. Foguem, C.; Manckoundia, P. Lewy body disease: Clinical and pathological “overlap syndrome” between synucleinopathies (Parkinson disease) and tauopathies (Alzheimer disease). Curr. Neurol. Neurosci. Rep. 2018, 18, 24. [Google Scholar] [CrossRef] [PubMed]
  15. Surendranathan, A.; Kane, J.P.M.; Bentley, A.; Barker, S.A.H.; Taylor, J.; Thomas, A.J.; Allan, L.M.; McNally, R.J.; James, P.W.; McKeith, I.G.; et al. Clinical diagnosis of Lewy body dementia. BJPsych Open 2020, 6, e61. [Google Scholar] [CrossRef] [PubMed]
  16. Chin, K.S.; Teodorczuk, A.; Watson, R. Dementia with Lewy bodies: Challenges in the diagnosis and management. Aust. N. Z. J. Psychiatry 2019, 53, 291–303. [Google Scholar] [CrossRef] [PubMed]
  17. Bir, S.C.; Khan, M.W.; Javalkar, V.; Toledo, E.G.; Kelley, R.E. Emerging Concepts in Vascular Dementia: A Review. J. Stroke Cerebrovasc. Dis. 2021, 30, 105864. [Google Scholar] [CrossRef]
  18. Hu, X.; De Silva, T.M.; Chen, J.; Faraci, F.M. Cerebral vascular disease and neurovascular injury in ischemic stroke. Circ. Res. 2017, 120, 449–471. [Google Scholar] [CrossRef] [PubMed]
  19. Hu, S.; Lan, T.; Wang, Y.; Ren, L. Individual Insomnia Symptom and Increased Hazard Risk of Cardiocerebral Vascular Diseases: A Meta-Analysis. Front. Psychiatry 2021, 12, 654719. [Google Scholar] [CrossRef]
  20. Maclin, J.M.A.; Wang, T.; Xiao, S. Biomarkers for the diagnosis of Alzheimer’s disease, dementia Lewy body, frontotemporal dementia and vascular dementia. Gen. Psychiatry 2019, 32, e100054. [Google Scholar] [CrossRef] [PubMed]
  21. Thijssen, E.H.; Verberk, I.M.W.; Kindermans, J.; Abramian, A.; Vanbrabant, J.; Ball, A.J.; Pijnenburg, Y.; Lemstra, A.W.; Flier, W.M.; Stoops, E.; et al. Differential diagnostic performance of a panel of plasma biomarkers for different types of dementia. Alzheimer’s Dement. 2022, 14, e12285. [Google Scholar] [CrossRef] [PubMed]
  22. Tang, L.; Wang, Y.; Xiang, J.; Yang, D.; Zhang, Y.; Xiang, Q.; Li, J. lncRNA and circRNA expression profiles in the hippocampus of Abeta(25-35)-induced AD mice treated with Tripterygium glycoside. Exp. Ther. Med. 2023, 26, 426. [Google Scholar] [CrossRef]
  23. Ramakrishna, S.; Muddashetty, R.S. Emerging Role of microRNAs in Dementia. J. Mol. Biol. 2019, 431, 743–1762. [Google Scholar] [CrossRef] [PubMed]
  24. Kim, W.R.; Park, E.G.; Lee, D.H.; Lee, Y.J.; Bae, W.H.; Kim, H.S. The Tumorigenic Role of Circular RNA-MicroRNA Axis in Cancer. Int. J. Mol. Sci. 2023, 24, 3050. [Google Scholar] [CrossRef] [PubMed]
  25. Ying, S.Y.; Chang, D.C.; Lin, S.L. The microRNA (miRNA): Overview of the RNA genes that modulate gene function. Mol. Biotechnol. 2008, 38, 257–268. [Google Scholar] [CrossRef] [PubMed]
  26. Leitão, A.L.; Enguita, F.J. A Structural View of miRNA Biogenesis and Function. Non-Coding RNA 2022, 8, 10. [Google Scholar] [CrossRef]
  27. Wahid, F.; Shehzad, A.; Khan, T.; Kim, Y.Y. MicroRNAs: Synthesis, mechanism, function, and recent clinical trials. Biochim. Biophys. Acta Mol. Cell Res. 2010, 1803, 1231–1243. [Google Scholar] [CrossRef] [PubMed]
  28. Lee, Y.; Ahn, C.; Han, J.; Choi, H.; Kim, J.; Yim, J.; Lee, J.; Provost, P.; Rådmark, O.; Kim, S.; et al. The nuclear RNase III Drosha initiates microRNA processing. Nature 2003, 425, 415–419. [Google Scholar] [CrossRef]
  29. Lee, Y.; Kim, M.; Han, J.; Yeom, K.H.; Lee, S.; Baek, S.H.; Kim, V.N. MicroRNA genes are transcribed by RNA polymerase II. EMBO J. 2004, 23, 4051–4060. [Google Scholar] [CrossRef]
  30. Lund, E.; Dahlberg, J.E. Substrate selectivity of exportin 5 and Dicer in the biogenesis of microRNAs. In Cold Spring Harbor Symposia on Quantitative Biology; Cold Spring Harbor Laboratory Press: Long Island, NY, USA, 2006; Volume 71, pp. 59–66. [Google Scholar]
  31. Pratt, A.J.; MacRae, I.J. The RNA-induced silencing complex: A versatile gene-silencing machine. J. Biol. Chem. 2009, 284, 17897–17901. [Google Scholar] [CrossRef]
  32. Tan, P.H.; Yang, L.C.; Ji, R.R. Therapeutic potential of RNA interference in pain medicine. Open Pain J. 2009, 2, 57. [Google Scholar] [CrossRef] [PubMed]
  33. Lin, S.; Gregory, R.I. MicroRNA biogenesis pathways in cancer. Nat. Rev. Cancer 2015, 15, 321–333. [Google Scholar] [CrossRef] [PubMed]
  34. Afonso-Grunz, F.; Müller, S. Principles of miRNA–mRNA interactions: Beyond sequence complementarity. Cell. Mol. Life Sci. 2015, 72, 3127–3141. [Google Scholar] [CrossRef] [PubMed]
  35. Grammatikakis, I.; Lal, A. Significance of lncRNA abundance to function. Mamm. Genome 2022, 33, 271–280. [Google Scholar] [CrossRef] [PubMed]
  36. Huaying, C.; Xing, J.; Luya, J.; Linhui, N.; Di, S.; Xianjun, D. A Signature of Five Long Non-Coding RNAs for Predicting the Prognosis of Alzheimer’s Disease Based on Competing Endogenous RNA Networks. Front. Aging Neurosci. 2020, 12, 598606. [Google Scholar] [CrossRef] [PubMed]
  37. Chen, L.L. Towards higher-resolution and in vivo understanding of lncRNA biogenesis and function. Nat. Methods 2022, 19, 1152–1155. [Google Scholar] [CrossRef] [PubMed]
  38. Nojima, T.; Proudfoot, N.J. Mechanisms of lncRNA biogenesis as revealed by nascent transcriptomics. Nat. Rev. Mol. Cell Biol. 2022, 23, 389–406. [Google Scholar] [CrossRef] [PubMed]
  39. Dahariya, S.; Paddibhatla, I.; Kumar, S.; Raghuwanshi, S.; Pallepati, A.; Gutti, R.K. Long non-coding RNA: Classification, biogenesis and functions in blood cells. Mol. Immunol. 2019, 112, 82–92. [Google Scholar] [CrossRef] [PubMed]
  40. Zhou, W.Y.; Cai, Z.R.; Liu, J.; Wang, D.S.; Ju, H.Q.; Xu, R.H. Circular RNA: Metabolism, functions and interactions with proteins. Mol. Cancer 2020, 19, 172. [Google Scholar] [CrossRef] [PubMed]
  41. Kristensen, L.S.; Andersen, M.S.; Stagsted, L.V.; Ebbesen, K.K.; Hansen, T.B.; Kjems, J. The biogenesis, biology and characterization of circular RNAs. Nat. Rev. Genet. 2019, 20, 675–691. [Google Scholar] [CrossRef] [PubMed]
  42. Greene, J.; Baird, A.M.; Brady, L.; Lim, M.; Gray, S.G.; McDermott, R.; Finn, S.P. Circular RNAs: Biogenesis, function and role in human diseases. Front. Mol. Biosci. 2017, 4, 38. [Google Scholar] [CrossRef] [PubMed]
  43. Dragomir, M.; Calin, G.A. Circular RNAs in cancer–lessons learned from microRNAs. Front. Oncol. 2018, 8, 179. [Google Scholar] [CrossRef] [PubMed]
  44. Chen, C.; Fang, F. hsa_circ_0006916 Exerts Effect on Amyloid Beta-Induced Neuron Injury by Targeting miR-217/HOMER1. Ann. Clin. Lab. Sci. 2023, 53, 181–191. [Google Scholar] [PubMed]
  45. Moreno-García, L.; López-Royo, T.; Calvo, A.C.; Toivonen, J.M.; de la Torre, M.; Moreno-Martínez, L.; Molina, N.; Aparicio, P.; Zaragoza, P.; Manzano, R.; et al. Competing Endogenous RNA Networks as Biomarkers in Neurodegenerative Diseases. Int. J. Mol. Sci. 2020, 21, 9582. [Google Scholar] [CrossRef] [PubMed]
  46. Ala, U. Competing Endogenous RNAs, Non-Coding RNAs and Diseases: An Intertwined Story. Cells 2020, 9, 1574. [Google Scholar] [CrossRef] [PubMed]
  47. Liu, X.; Liu, H.; Jia, X.; He, R.; Zhang, X.; Zhang, W. Changing Expression Profiles of Messenger RNA, MicroRNA, Long Non-coding RNA, and Circular RNA Reveal the Key Regulators and Interaction Networks of Competing Endogenous RNA in Pulmonary Fibrosis. Front. Genet. 2020, 11, 558095. [Google Scholar] [CrossRef] [PubMed]
  48. Lee, Y.J.; Kim, W.R.; Park, E.G.; Lee, D.H.; Kim, J.M.; Shin, H.J.; Jeong, H.S.; Roh, H.Y.; Kim, H.S. Exploring the Key Signaling Pathways and ncRNAs in Colorectal Cancer. Int. J. Mol. Sci. 2024, 25, 4548. [Google Scholar] [CrossRef]
  49. Fiannaca, A.; Paglia, L.L.; Rosa, M.L.; Rizzo, R.; Urso, A. miRTissue ce: Extending miRTissue web service with the analysis of ceRNA-ceRNA interactions. BMC Bioinform. 2020, 21, 199. [Google Scholar] [CrossRef] [PubMed]
  50. Asadi, M.R.; Abed, S.; Kouchakali, G.; Fattahi, F.; Sabaie, H.; Moslehian, M.S.; Sharifi-Bonab, M.; Hussen, B.M.; Taheri, M.; Ghafouri-Fard, S.; et al. Competing endogenous RNA (ceRNA) networks in Parkinson’s disease: A systematic review. Front. Cell. Neurosci. 2023, 17, 1044634. [Google Scholar] [CrossRef]
  51. Asanomi, Y.; Shigemizu, D.; Akiyama, S.; Sakurai, T.; Ozaki, K.; Ochiya, T.; Niida, S. Dementia subtype prediction models constructed by penalized regression methods for multiclass classification using serum microRNA expression data. Sci. Rep. 2021, 11, 20947. [Google Scholar] [CrossRef] [PubMed]
  52. Koriath, C.; Kenny, J.; Adamson, G.; Druyeh, R.; Taylor, W.; Beck, J.; Quinn, L.; Mok, T.H.; Dimitriadis, A.; Norsworthy, P.; et al. Predictors for a dementia gene mutation based on gene-panel next-generation sequencing of a large dementia referral series. Mol. Psychiatry 2020, 25, 3399–3412. [Google Scholar] [CrossRef] [PubMed]
  53. Armstrong, R.A. Risk factors for Alzheimer’s disease. Folia Neuropathol. 2019, 57, 87–105. [Google Scholar] [CrossRef]
  54. Liu, S.; Fan, M.; Zheng, Q.; Hao, S.; Yang, L.; Xia, Q.; Qi, C.; Ge, J. MicroRNAs in Alzheimer’s disease: Potential diagnostic markers and therapeutic targets. Biomed. Pharmacother. 2022, 148, 112681. [Google Scholar] [CrossRef] [PubMed]
  55. Wang, L.; Shui, X.; Diao, Y.; Chen, D.; Zhou, Y.; Lee, T.H. Potential Implications of miRNAs in the Pathogenesis, Diagnosis, and Therapeutics of Alzheimer’s Disease. Int. J. Mol. Sci. 2023, 24, 16259. [Google Scholar] [CrossRef] [PubMed]
  56. Sun, C.; Liu, J.; Duan, F.; Cong, L.; Qi, X. The role of the microRNA regulatory network in Alzheimer’s disease: A bioinformatics analysis. Arch. Med. Sci. 2022, 18, 206–222. [Google Scholar] [CrossRef] [PubMed]
  57. Wang, R.; Chopra, N.; Nho, K.; Maloney, B.; Obukhov, A.G.; Nelson, P.T.; Counts, E.; Lahiri, D.K. Human microRNA (miR-20b-5p) modulates Alzheimer’s disease pathways and neuronal function, and a specific polymorphism close to the MIR20B gene influences Alzheimer’s biomarkers. Mol. Psychiatry 2022, 27, 1256–1273. [Google Scholar] [CrossRef] [PubMed]
  58. Liang, C.; Mu, Y.; Tian, H.; Wang, D.; Zhang, S.; Wang, H.; Liu, Y.; Di, C. MicroRNA-140 silencing represses the incidence of Alzheimer’s disease. Neurosci. Lett. 2021, 758, 135674. [Google Scholar] [CrossRef] [PubMed]
  59. 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. 2020, 127, 339–346. [Google Scholar] [CrossRef] [PubMed]
  60. Ruan, Z.; Li, Y.; He, R.; Li, X. Inhibition of microRNA-10b-5p up-regulates HOXD10 to attenuate Alzheimer’s disease in rats via the Rho/ROCK signalling pathway. J. Drug Target. 2021, 29, 531–540. [Google Scholar] [CrossRef] [PubMed]
  61. Xiao, G.; Chen, Q.; Zhang, X. MicroRNA-455-5p/CPEB1 pathway mediates Abeta-related learning and memory deficits in a mouse model of Alzheimer’s disease. Brain Res. Bull. 2021, 177, 282–294. [Google Scholar] [CrossRef] [PubMed]
  62. Wu, G.D.; Li, Z.H.; Li, X.; Zheng, T.; Zhang, D.K. microRNA-592 blockade inhibits oxidative stress injury in Alzheimer’s disease astrocytes via the KIAA0319-mediated Keap1/Nrf2/ARE signaling pathway. Exp. Neurol. 2020, 324, 113128. [Google Scholar] [CrossRef]
  63. Kim, S.H.; Choi, K.Y.; Park, Y.; McLean, C.; Park, J.; Lee, J.H.; Kim, B.C.; Huh, Y.H.; Lee, K.H.; Song, W.K. Enhanced Expression of microRNA-1273g-3p Contributes to Alzheimer’s Disease Pathogenesis by Regulating the Expression of Mitochondrial Genes. Cells 2021, 10, 2697. [Google Scholar] [CrossRef] [PubMed]
  64. Koh, H.S.; Lee, S.; Lee, H.J.; Min, J.W.; Iwatsubo, T.; Teunissen, C.E.; Cho, H.J.; Ryu, J.H. Targeting MicroRNA-485-3p Blocks Alzheimer’s Disease Progression. Int. J. Mol. Sci. 2022, 23, 3566. [Google Scholar] [CrossRef]
  65. 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] [PubMed]
  66. Li, Y.; Meng, S.; Di, W.; Xia, M.; Dong, L.; Zhao, Y.; Ling, S.; He, J.; Xue, X.; Chen, X.; et al. Amyloid-beta protein and MicroRNA-384 in NCAM-Labeled exosomes from peripheral blood are potential diagnostic markers for Alzheimer’s disease. CNS Neurosci. Ther. 2022, 28, 1093–1107. [Google Scholar] [CrossRef] [PubMed]
  67. Imperatore, J.A.; Then, M.L.; McDougal, K.B.; Mihailescu, M.R. Characterization of a G-Quadruplex Structure in Pre-miRNA-1229 and in Its Alzheimer’s Disease-Associated Variant rs2291418: Implications for miRNA-1229 Maturation. Int. J. Mol. Sci. 2020, 21, 767. [Google Scholar] [CrossRef] [PubMed]
  68. Pena-Bautista, C.; Tarazona-Sanchez, A.; Braza-Boils, A.; Balaguer, A.; Ferre-Gonzalez, L.; Canada-Martinez, A.J.; Baquero, M.; Chafer-Pericas, C. Plasma microRNAs as potential biomarkers in early Alzheimer disease expression. Sci. Rep. 2022, 12, 15589. [Google Scholar] [CrossRef] [PubMed]
  69. Sørensen, S.S.; Nygaard, A.B.; Christensen, T. miRNA expression profiles in cerebrospinal fluid and blood of patients with Alzheimer’s disease and other types of dementia–an exploratory study. Transl. Neurodegener. 2016, 5, 6. [Google Scholar] [CrossRef] [PubMed]
  70. Sarkar, S.; Jun, S.; Rellick, S.; Quintana, D.D.; Cavendish, J.Z.; Simpkins, J.W. Expression of microRNA-34a in Alzheimer’s disease brain targets genes linked to synaptic plasticity, energy metabolism, and resting state network activity. Brain Res. 2016, 1646, 139–151. [Google Scholar] [CrossRef] [PubMed]
  71. Cong, L.; Cong, Y.; Feng, N.; Liang, W.; Wu, Y. Up-regulated microRNA-132 reduces the cognition-damaging effect of sevoflurane on Alzheimer’s disease rats by inhibiting FOXA1. Genomics 2021, 113, 3644–3652. [Google Scholar] [CrossRef]
  72. He, B.; Chen, W.; Zeng, J.; Tong, W.; Zheng, P. MicroRNA-326 decreases tau phosphorylation and neuron apoptosis through inhibition of the JNK signaling pathway by targeting VAV1 in Alzheimer’s disease. J. Cell. Physiol. 2020, 235, 14. [Google Scholar] [CrossRef] [PubMed]
  73. Liang, C.; Zou, T.; Zhang, M.; Fan, W.; Zhang, T.; Jiang, Y.; Cai, Y.; Chen, F.; Chen, X.; Sun, Y.; et al. MicroRNA-146a switches microglial phenotypes to resist the pathological processes and cognitive degradation of Alzheimer’s disease. Theranostics 2021, 11, 4103–4121. [Google Scholar] [CrossRef] [PubMed]
  74. Cao, J.; Huang, M.; Guo, L.; Zhu, L.; Hou, J.; Zhang, L.; Pero, A.; Ng, S.; Gaamouch, F.E.; Elder, G.; et al. MicroRNA-195 rescues ApoE4-induced cognitive deficits and lysosomal defects in Alzheimer’s disease pathogenesis. Mol. Psychiatry 2021, 26, 4687–4701. [Google Scholar] [CrossRef] [PubMed]
  75. Wang, J.; Chen, C.; Zhang, Y. An investigation of microRNA-103 and microRNA-107 as potential blood-based biomarkers for disease risk and progression of Alzheimer’s disease. J. Clin. Lab. Anal. 2020, 34, e23006. [Google Scholar] [CrossRef] [PubMed]
  76. Kumar, S.; Morton, H.; Sawant, N.; Orlov, E.; Bunquin, L.E.; Pradeepkiran, J.A.; Alvir, R.; Reddy, P.H. MicroRNA-455-3p improves synaptic, cognitive functions and extends lifespan: Relevance to Alzheimer’s disease. Redox Biol. 2021, 48, 102182. [Google Scholar] [CrossRef] [PubMed]
  77. Nakano, M.; Kubota, K.; Kobayashi, E.; Chikenji, T.S.; Saito, Y.; Konari, N.; Fujimiya, M. Bone marrow-derived mesenchymal stem cells improve cognitive impairment in an Alzheimer’s disease model by increasing the expression of microRNA-146a in hippocampus. Sci. Rep. 2020, 10, 10772. [Google Scholar] [CrossRef] [PubMed]
  78. Du, W.; Lei, C.; Dong, Y. MicroRNA-149 is downregulated in Alzheimer’s disease and inhibits β-amyloid accumulation and ameliorates neuronal viability through targeting BACE1. Genet. Mol. Biol. 2021, 44, e20200064. [Google Scholar] [CrossRef] [PubMed]
  79. Souza, V.C.; Morais, G.S., Jr.; Henriques, A.D.; Machado-Silva, W.; Perez, D.I.V.; Brito, C.J.; Camargos, E.F.; Moraes, C.F.; Nóbrega, O.T. Whole-Blood Levels of MicroRNA-9 Are Decreased in Patients With Late-Onset Alzheimer Disease. Am. J. Alzheimer’s Dis. Other Demen. 2020, 35, 1533317520911573. [Google Scholar] [CrossRef] [PubMed]
  80. Chen, W.; Wu, L.; Hu, Y.; Jiang, L.; Liang, N.; Chen, J.; Qin, H.; Tang, N. MicroRNA-107 Ameliorates Damage in a Cell Model of Alzheimer’s Disease by Mediating the FGF7/FGFR2/PI3K/Akt Pathway. J. Mol. Neurosci. 2020, 70, 1589–1597. [Google Scholar] [CrossRef] [PubMed]
  81. Pan, K.; Chen, S.; Wang, Y.; Yao, W.; Gao, X. MicroRNA-23b attenuates tau pathology and inhibits oxidative stress by targeting GnT-III in Alzheimer’s disease. Neuropharmacology 2021, 196, 108671. [Google Scholar] [CrossRef] [PubMed]
  82. Sha, S.; Shen, X.; Cao, Y.; Qu, L. Mesenchymal stem cells-derived extracellular vesicles ameliorate Alzheimer’s disease in rat models via the microRNA-29c-3p/BACE1 axis and the Wnt/beta-catenin pathway. Aging 2021, 13, 15285–15306. [Google Scholar] [CrossRef] [PubMed]
  83. Barros-Viegas, A.T.; Carmona, V.; Ferreiro, E.; Guedes, J.; Cardoso, A.M.; Cunha, P.; Almeida, L.P.; Oliveira, C.R.; Magalhães, J.P.; Peça, J.; et al. miRNA-31 Improves Cognition and Abolishes Amyloid-beta Pathology by Targeting APP and BACE1 in an Animal Model of Alzheimer’s Disease. Mol. Ther. Nucleic Acids 2020, 19, 1219–1236. [Google Scholar] [CrossRef] [PubMed]
  84. Wang, Y.; Chang, Q. MicroRNA miR-212 regulates PDCD4 to attenuate Aβ25–35-induced neurotoxicity via PI3KAKT signaling pathway in Alzheimer’s disease. Biotechnol. Lett. 2020, 42, 1789–1797. [Google Scholar] [CrossRef] [PubMed]
  85. Xia, P.; Chen, J.; Liu, Y.; Cui, X.; Wang, C.; Zong, S.; Wang, L.; Lu, Z. MicroRNA-22-3p ameliorates Alzheimer’s disease by targeting SOX9 through the NF-κB signaling pathway in the hippocampus. J. Neuroinflamm. 2022, 19, 180. [Google Scholar] [CrossRef] [PubMed]
  86. Xue, B.; Qu, Y.; Zhang, X.; Xu, X.F. miRNA-126a-3p participates in hippocampal memory via alzheimer’s disease-related proteins. Cereb. Cortex 2022, 32, 4763–4781. [Google Scholar] [CrossRef] [PubMed]
  87. Zhai, L.; Shen, H.; Sheng, Y.; Guan, Q. ADMSC Exo-MicroRNA-22 improve neurological function and neuroinflammation in mice with Alzheimer’s disease. J. Cell. Mol. Med. 2021, 25, 7513–7523. [Google Scholar] [CrossRef] [PubMed]
  88. Piscopo, P.; Grasso, M.; Manzini, V.; Zeni, A.; Castelluzzo, M.; Fontana, F.; Talarico, G.; Castellano, A.E.; Rivabene, R.; Crestini, A.; et al. Identification of miRNAs regulating MAPT expression and their analysis in plasma of patients with dementia. Front. Mol. Neurosci. 2023, 16, 1127163. [Google Scholar] [CrossRef]
  89. Smith, P.Y.; Hernandez-Rapp, J.; Jolivette, F.; Lecours, C.; Bisht, K.; Goupil, C.; Dorval, V.; Parsi, S.; Morin, F.; Planel, E.; et al. miR-132/212 deficiency impairs tau metabolism and promotes pathological aggregation in vivo. Hum. Mol. Genet. 2015, 24, 6721–6735. [Google Scholar] [CrossRef] [PubMed]
  90. Zhang, J.; Hu, M.; Teng, Z.; Tang, Y.P.; Chen, C. Synaptic and cognitive improvements by inhibition of 2-AG metabolism are through upregulation of microRNA-188-3p in a mouse model of Alzheimer’s disease. J. Neurosci. 2014, 34, 14919–14933. [Google Scholar] [CrossRef] [PubMed]
  91. Lau, P.; Bossers, K.; Janky, R.S.; Salta, E.; Frigerio, C.S.; Barbash, S.; Rothman, R.; Sierksma, A.S.R.; Thathiah, A.; Greenberg, D.; et al. Alteration of the micro RNA network during the progression of Alzheimer’s disease. EMBO Mol. Med. 2013, 5, 1613–1634. [Google Scholar] [CrossRef]
  92. Mezache, L.; Mikhail, M.; Garofalo, M.; Nuovo, G.J. Reduced miR-512 and the elevated expression of its targets cFLIP and MCL1 localize to neurons with hyperphosphorylated tau protein in Alzheimer disease. Appl. Immunohistochem. Mol. Morphol. 2015, 23, 615–623. [Google Scholar] [CrossRef] [PubMed]
  93. Hampel, H.; Vassar, R.; De Strooper, B.; Hardy, J.; Willem, M.; Singh, N.; Zhou, J.; Yan, R.; Vanmechelen, E.; Vos, A.D.; et al. The beta-Secretase BACE1 in Alzheimer’s Disease. Biol. Psychiatry 2021, 89, 745–756. [Google Scholar] [CrossRef] [PubMed]
  94. Cui, G.H.; Zhu, J.; Wang, Y.C.; Wu, J.; Liu, J.R.; Guo, H.D. Effects of exosomal miRNAs in the diagnosis and treatment of Alzheimer’s disease. Mech. Ageing Dev. 2021, 200, 111593. [Google Scholar] [CrossRef] [PubMed]
  95. Gurunathan, S.; Kang, M.H.; Jeyaraj, M.; Qasim, M.; Kim, J.H. Review of the isolation, characterization, biological function, and multifarious therapeutic approaches of exosomes. Cells 2019, 8, 307. [Google Scholar] [CrossRef] [PubMed]
  96. Zheng, D.; Huo, M.; Li, B.; Wang, W.; Piao, H.; Wang, Y.; Zhu, Z.; Li, D.; Wang, T.; Liu, K. The Role of Exosomes and Exosomal MicroRNA in Cardiovascular Disease. Front. Cell Dev. Biol. 2020, 8, 616161. [Google Scholar] [CrossRef] [PubMed]
  97. Yoon, Y.J.; Kim, O.Y.; Gho, Y.S. Extracellular vesicles as emerging intercellular communicasomes. BMB Rep. 2014, 47, 531. [Google Scholar] [CrossRef] [PubMed]
  98. Liu, C.G.; Shuang, M.E.N.G.; Ying, L.I.; Yao, L.U.; Yue, Z.H.A.O.; Wang, P.C. MicroRNA-135a in ABCA1-labeled Exosome is a Serum Biomarker Candidate for Alzheimer’s Disease. Biomed. Environ. Sci. 2021, 34, 19–28. [Google Scholar] [PubMed]
  99. Manna, I.; De Benedittis, S.; Quattrone, A.; Maisano, D.; Iaccino, E.; Quattrone, A. Exosomal miRNAs as potential diagnostic biomarkers in Alzheimer’s disease. Pharmaceuticals 2020, 13, 243. [Google Scholar] [CrossRef] [PubMed]
  100. Chen, J.J.; Zhao, B.; Zhao, J.; Li, S. Potential roles of exosomal microRNAs as diagnostic biomarkers and therapeutic application in Alzheimer’s disease. Neural Plast. 2017, 2017, 7027380. [Google Scholar] [CrossRef] [PubMed]
  101. Parker, M.; Barlow, S.; Hoe, J.; Aitken, L. Persistent barriers and facilitators to seeking help for a dementia diagnosis: A systematic review of 30 years of the perspectives of carers and people with dementia. Int. Psychogeriatr. 2020, 32, 611–634. [Google Scholar] [CrossRef] [PubMed]
  102. Pemberton, H.G.; Zaki, L.A.; Goodkin, O.; Das, R.K.; Steketee, R.M.; Barkhof, F.; Vernooij, M.W. Correction to: Technical and clinical validation of commercial automated volumetric MRI tools for dementia diagnosis-a systematic review. Neuroradiology 2021, 63, 1955. [Google Scholar] [CrossRef] [PubMed]
  103. Raji, C.A.; Benzinger, T.L. The Value of Neuroimaging in Dementia Diagnosis. Continuum 2022, 28, 800–821. [Google Scholar] [CrossRef] [PubMed]
  104. Kosiol, N.; Juranek, S.; Brossart, P.; Heine, A.; Paeschke, K. G-quadruplexes: A promising target for cancer therapy. Mol. Cancer 2021, 20, 1–18. [Google Scholar] [CrossRef] [PubMed]
  105. Bidzinska, J.; Cimino-Reale, G.; Zaffaroni, N.; Folini, M. G-quadruplex structures in the human genome as novel therapeutic targets. Molecules 2013, 18, 12368–12395. [Google Scholar] [CrossRef] [PubMed]
  106. Zhou, H.; Yang, C.; Chen, H.; Li, X.; Li, Y.; Fan, X. A simple G-quadruplex molecular beacon-based biosensor for highly selective detection of microRNA. Biosens. Bioelectron. 2017, 87, 552–557. [Google Scholar] [CrossRef] [PubMed]
  107. Karantzoulis, S.; Galvin, J.E. Distinguishing Alzheimer’s disease from other major forms of dementia. Expert Rev. Neurother. 2011, 11, 1579–1591. [Google Scholar] [CrossRef] [PubMed]
  108. Goodman, R.A.; Lochner, K.A.; Thambisetty, M.; Wingo, T.S.; Posner, S.F.; Ling, S.M. Prevalence of dementia subtypes in United States Medicare fee-for-service beneficiaries, 2011–2013. Alzheimer’s Dement. 2017, 13, 28–37. [Google Scholar] [CrossRef] [PubMed]
  109. Iacono, D.; Raiciulescu, S.; Olsen, C.; Perl, D.P. Traumatic Brain Injury Exposure Lowers Age of Cognitive Decline in AD and Non-AD Conditions. Front. Neurol. 2021, 12, 573401. [Google Scholar] [CrossRef] [PubMed]
  110. Jiao, J.; Herl, L.D.; Farese, R.V., Jr.; Gao, F.B. MicroRNA-29b regulates the expression level of human progranulin, a secreted glycoprotein implicated in frontotemporal dementia. PLoS ONE 2010, 5, e10551. [Google Scholar] [CrossRef] [PubMed]
  111. Piscopo, P.; Grasso, M.; Fontana, F.; Crestini, A.; Puopolo, M.; Del Vescovo, V.; Venerosi, A.; Calamandrei, G.; Vencken, S.F.; Greene, C.M.; et al. Reduced miR-659-3p Levels Correlate with Progranulin Increase in Hypoxic Conditions: Implications for Frontotemporal Dementia. Front. Mol. Neurosci. 2016, 9, 31. [Google Scholar] [CrossRef] [PubMed]
  112. Schneider, R.; McKeever, P.; Kim, T.; Graff, C.; van Swieten, J.C.; Karydas, A.; Boxer, A.; Rosen, H.; Miller, B.L.; Laforce, R., Jr.; et al. Downregulation of exosomal miR-204-5p and miR-632 as a biomarker for FTD: A GENFI study. J. Neurol. Neurosurg. Psychiatry 2018, 89, 851–858. [Google Scholar] [CrossRef] [PubMed]
  113. Chen-Plotkin, A.S.; Unger, T.L.; Gallagher, M.D.; Bill, E.; Kwong, L.K.; Volpicelli-Daley, L.; Busch, J.I.; Akle, S.; Grossman, M.; Deerlin, V.V.; et al. TMEM106B, the Risk Gene for Frontotemporal Dementia, Is Regulated by the microRNA-132/212 Cluster and Affects Progranulin Pathways. J. Neurosci. 2012, 32, 11213–11227. [Google Scholar] [CrossRef] [PubMed]
  114. Gascon, E.; Lynch, K.; Ruan, H.; Almeida, S.; Verheyden, J.M.; Seeley, W.W.; Dickson, D.W.; Petrucelli, L.; Sun, D.; Jiao, J.; et al. Alterations in microRNA-124 and AMPA receptors contribute to social behavioral deficits in frontotemporal dementia. Nat. Med. 2014, 20, 1444–1451. [Google Scholar] [CrossRef] [PubMed]
  115. Piscopo, P.; Grasso, M.; Puopolo, M.; D’Acunto, E.; Talarico, G.; Crestini, A.; Gasparini, M.; Campopiano, R.; Gambardella, S.; Castellano, A.E.; et al. Circulating miR-127-3p as a Potential Biomarker for Differential Diagnosis in Frontotemporal Dementia. J. Alzheimer’s Dis. 2018, 65, 455–464. [Google Scholar] [CrossRef] [PubMed]
  116. Tagliafierro, L.; Glenn, O.C.; Zamora, M.E.; Beach, T.G.; Woltjer, R.L.; Lutz, M.W.; Chiba-Falek, O. Genetic analysis of SNCA 3’UTR and its corresponding miRNAs in relation to Parkinson’s compared to Dementia with Lewy Bodies. Alzheimer’s. Dement. 2017, 13, 1237. [Google Scholar] [CrossRef] [PubMed]
  117. Ren, Z.; Yu, J.; Wu, Z.; Si, W.; Li, X.; Liu, Y.; Zhou, J.; Deng, R.; Chen, D. MicroRNA-210-5p Contributes to Cognitive Impairment in Early Vascular Dementia Rat Model Through Targeting Snap25. Front. Mol. Neurosci. 2018, 11, 388. [Google Scholar] [CrossRef] [PubMed]
  118. Liu, X.; Zhang, R.; Wu, Z.; Si, W.; Ren, Z.; Zhang, S.; Zhou, J.; Chen, D. miR-134-5p/Foxp2/Syn1 is involved in cognitive impairment in an early vascular dementia rat model. Int. J. Mol. Med. 2019, 44, 1729–1740. [Google Scholar] [CrossRef] [PubMed]
  119. Wang, L.; Yang, J.W.; Lin, L.T.; Huang, J.; Wang, X.R.; Su, X.T.; Cao, Y.; Fisher, M.; Liu, C.Z. Acupuncture Attenuates Inflammation in Microglia of Vascular Dementia Rats by Inhibiting miR-93-Mediated TLR4/MyD88/NF-kappaB Signaling Pathway. Oxid. Med. Cell. Longev. 2020, 2020, 8253904. [Google Scholar] [PubMed]
  120. Wei, C.; Xu, X.; Zhu, H.; Zhang, X.; Gao, Z. Promotive role of microRNA-150 in hippocampal neurons apoptosis in vascular dementia model rats. Mol. Med. Rep. 2021, 23, 257. [Google Scholar] [CrossRef] [PubMed]
  121. Xia, H.; Wang, H.; Li, Y.; Luo, Y.; Liu, Y.; Wang, F. Study on the Function of miR-134 on Cognitive Function of Vascular Dementia (VD) Rats and Mechanism about Oxidative Stress and Autophagy and Cofilin 2 Level. J. Biomater. Tiss. Eng. 2022, 12, 1994–2000. [Google Scholar] [CrossRef]
  122. Han, X.; Zhou, L.; Tu, Y.; Wei, J.; Zhang, J.; Jiang, G.; Shi, Q.; Ying, H. Circulating exo-miR-154-5p regulates vascular dementia through endothelial progenitor cell-mediated angiogenesis. Front. Cell. Neurosci. 2022, 16, 881175. [Google Scholar] [CrossRef] [PubMed]
  123. Li, X.; Chen, S.; Wang, S.; Bai, X.; Lv, P. miR-181a regulates mitophagy and improves cognitive function in vascular dementia via targeting PINK1/Parkin pathway. Mater. Express 2023, 13, 1234–1240. [Google Scholar] [CrossRef]
  124. Qian, X.; Xu, Q.; Li, G.; Bu, Y.; Sun, F.; Zhang, J. Therapeutic Effect of Idebenone on Rats with Vascular Dementia via the MicroRNA-216a/RSK2/NF-kappaB Axis. Neuropsychiatr. Dis. Treat. 2021, 17, 533–543. [Google Scholar] [CrossRef]
  125. Ma, X.; Wang, Y.; Shi, Y.; Li, S.; Liu, J.; Li, X.; Zhong, W.; Pan, Q. Exosomal miR-132-3p from mesenchymal stromal cells improves synaptic dysfunction and cognitive decline in vascular dementia. Stem Cell Res. Ther. 2022, 13, 315. [Google Scholar] [CrossRef] [PubMed]
  126. Zheng, W.; Zhang, J.; Zhou, B.; Chang, H. MiR-322-5p Alleviates Cell Injury and Impairment of Cognitive Function in Vascular Dementia by Targeting TSPAN5. Yonsei Med. J. 2022, 63, 282–291. [Google Scholar] [CrossRef] [PubMed]
  127. Kirola, L.; Mukherjee, A.; Mutsuddi, M. Recent Updates on the Genetics of Amyotrophic Lateral Sclerosis and Frontotemporal Dementia. Mol. Neurobiol. 2022, 59, 5673–5694. [Google Scholar] [CrossRef] [PubMed]
  128. Matar, E.; Ehgoetz Martens, K.A.; Halliday, G.M.; Lewis, S.J. Clinical features of Lewy body dementia: Insights into diagnosis and pathophysiology. J. Neurol. 2020, 267, 380–389. [Google Scholar] [CrossRef] [PubMed]
  129. Martini, A.; Weis, L.; Schifano, R.; Pistonesi, F.; Fiorenzato, E.; Antonini, A.; Biundo, R. Differences in cognitive profiles between Lewy body and Parkinson’s disease dementia. J. Neural. Transm. 2020, 127, 323–330. [Google Scholar] [CrossRef] [PubMed]
  130. Wightman, D.P.; Savage, J.E.; Tissink, E.; Romero, C.; Jansen, I.E.; Posthuma, D. The genetic overlap between Alzheimer’s disease, amyotrophic lateral sclerosis, Lewy body dementia, and Parkinson’s disease. Neurobiol. Aging 2023, 127, 99–112. [Google Scholar] [CrossRef] [PubMed]
  131. Blauwendraat, C.; Reed, X.; Krohn, L.; Heilbron, K.; Bandres-Ciga, S.; Tan, M.; Gibbs, J.R.; Hernandez, D.G.; Kumaran, R.; Langston, R.; et al. Genetic modifiers of risk and age at onset in GBA associated Parkinson’s disease and Lewy body dementia. Brain 2020, 143, 234–248. [Google Scholar] [CrossRef] [PubMed]
  132. Sezgin, M.; Bilgic, B.; Tinaz, S.; Emre, M. Parkinson’s Disease Dementia and Lewy Body Disease. Semin. Neurol. 2019, 39, 274–282. [Google Scholar] [CrossRef] [PubMed]
  133. Marsal-García, L.; Urbizu, A.; Arnaldo, L.; Campdelacreu, J.; Vilas, D.; Ispierto, L.; Gascón-Bayarri, J.; Reñé, R.; Álvarez, R.; Beyer, K. Expression Levels of an Alpha-Synuclein Transcript in Blood May Distinguish between Early Dementia with Lewy Bodies and Parkinson’s Disease. Int. J. Mol. Sci. 2021, 22, 725. [Google Scholar] [CrossRef] [PubMed]
  134. Beishon, L.C.; Hosford, P.; Gurung, D.; Brassard, P.; Minhas, J.S.; Robinson, T.G.; Haunton, V.; Panerai, R.B. The role of the autonomic nervous system in cerebral blood flow regulation in dementia: A review. Auton. Neurosci. 2022, 240, 102985. [Google Scholar] [CrossRef] [PubMed]
  135. Prajjwal, P.; Marsool, M.D.M.; Inban, P.; Sharma, B.; Asharaf, S.; Aleti, S.; Gadam, S.; Sakini, A.S.A.; Hadi, D.D. Vascular dementia subtypes, pathophysiology, genetics, neuroimaging, biomarkers, and treatment updates along with its association with Alzheimer’s dementia and diabetes mellitus. Dis. Mon. 2023, 69, 101557. [Google Scholar] [CrossRef] [PubMed]
  136. Takeda, S.; Rakugi, H.; Morishita, R. Roles of vascular risk factors in the pathogenesis of dementia. Hypertens. Res. 2020, 43, 162–167. [Google Scholar] [CrossRef] [PubMed]
  137. Chen, S.; Zhang, Y.; Ding, X.; Li, W. Identification of lncRNA/circRNA-miRNA-mRNA ceRNA Network as Biomarkers for Hepatocellular Carcinoma. Front. Genet. 2022, 13, 838869. [Google Scholar] [CrossRef] [PubMed]
  138. Olufunmilayo, E.O.; Holsinger, R.D. Roles of non-coding RNA in alzheimer’s disease pathophysiology. Int. J. Mol. Sci. 2023, 24, 12498. [Google Scholar] [CrossRef] [PubMed]
  139. Wei, C.; Luo, T.; Zou, S.; Zhou, X.; Shen, W.; Ji, X.; Li, Q.; Wu, A. Differentially expressed lncRNAs and miRNAs with associated ceRNA networks in aged mice with postoperative cognitive dysfunction. Oncotarget 2017, 8, 55901. [Google Scholar] [CrossRef] [PubMed]
  140. Boniolo, F.; Hoffmann, M.; Roggendorf, N.; Tercan, B.; Baumbach, J.; Castro, M.A.; Robertson, A.G.; Saur, D.; List, M. spongEffects: ceRNA modules offer patient-specific insights into the miRNA regulatory landscape. Bioinformatics 2023, 39, btad276. [Google Scholar] [CrossRef] [PubMed]
  141. Guil, S.; Esteller, M. RNA–RNA interactions in gene regulation: The coding and noncoding players. Trends Biochem. Sci. 2015, 40, 248–256. [Google Scholar] [CrossRef] [PubMed]
  142. Chan, J.J.; Tay, Y. Noncoding RNA: RNA regulatory networks in cancer. Int. J. Mol. Sci. 2018, 19, 1310. [Google Scholar] [CrossRef] [PubMed]
  143. He, J.; Li, X.; Zhang, Y.; Zhang, Q.; Li, L. Comprehensive Analysis of ceRNA Regulation Network Involved in the Development of Coronary Artery Disease. Biomed Res. Int. 2021, 2021, 665815. [Google Scholar] [CrossRef] [PubMed]
  144. Lin, W.; Liu, H.; Tang, Y.; Wei, Y.; Wei, W.; Zhang, L.; Chen, J. The development and controversy of competitive endogenous RNA hypothesis in non-coding genes. Mol. Cell. Biochem. 2021, 476, 109–123. [Google Scholar] [CrossRef] [PubMed]
  145. Teixeira, L.C.R.; Mamede, I.; Luizon, M.R.; Gomes, K.B. Role of long non-coding RNAs in the pathophysiology of Alzheimer’s disease and other dementias. Mol. Biol. Rep. 2024, 51, 270. [Google Scholar] [CrossRef] [PubMed]
  146. Venkatesh, J.; Wasson, M.C.D.; Brown, J.M.; Fernando, W.; Marcato, P. LncRNA-miRNA axes in breast cancer: Novel points of interaction for strategic attack. Cancer Lett. 2021, 509, 81–88. [Google Scholar] [CrossRef] [PubMed]
  147. Ballantyne, M.D.; McDonald, R.A.; Baker, A.H. lncRNA/MicroRNA interactions in the vasculature. Clin. Pharm. Therap. 2016, 99, 494–501. [Google Scholar] [CrossRef] [PubMed]
  148. Entezari, M.; Taheriazam, A.; Orouei, S.; Fallah, S.; Sanaei, A.; Hejazi, E.S.; Kakavand, A.; Rezaei, S.; Heidari, H.; Behroozaghdam, M.; et al. LncRNA-miRNA axis in tumor progression and therapy response: An emphasis on molecular interactions and therapeutic interventions. Biomed. Pharmacother. 2022, 154, 113609. [Google Scholar] [CrossRef] [PubMed]
  149. Tehrani, S.S.; Ebrahimi, R.; Al-e-Ahmad, A.; Panahi, G.; Meshkani, R.; Younesi, S.; Saadat, P.; Parsian, H. Competing Endogenous RNAs (CeRNAs): Novel Network in Neurological Disorders. Curr. Med. Chem. 2021, 28, 5983–6010. [Google Scholar] [CrossRef] [PubMed]
  150. Sebastian-delaCruz, M.; Gonzalez-Moro, I.; Olazagoitia-Garmendia, A.; Castellanos-Rubio, A.; Santin, I. The role of lncRNAs in gene expression regulation through mRNA stabilization. Non-Coding RNA 2021, 7, 3. [Google Scholar] [CrossRef] [PubMed]
  151. Sabaie, H.; Amirinejad, N.; Asadi, M.R.; Jalaiei, A.; Daneshmandpour, Y.; Rezaei, O.; Taheri, M.; Rezazadeh, M. Molecular Insight Into the Therapeutic Potential of Long Non-coding RNA-Associated Competing Endogenous RNA Axes in Alzheimer’s Disease: A Systematic Scoping Review. Front. Aging Neurosci. 2021, 13, 742242. [Google Scholar] [CrossRef]
  152. Arun, G.; Aggarwal, D.; Spector, D.L. MALAT1 Long Non-Coding RNA: Functional Implications. Non-Coding RNA 2020, 6, 22. [Google Scholar] [CrossRef] [PubMed]
  153. Li, K.; Wang, Z. lncRNA NEAT1: Key player in neurodegenerative diseases. Ageing Res. Rev. 2023, 86, 101878. [Google Scholar] [CrossRef] [PubMed]
  154. Maniati, M.S.; Maniati, M.; Yousefi, T.; Ahmadi-Ahangar, A.; Tehrani, S.S. New insights into the role of microRNAs and long noncoding RNAs in most common neurodegenerative diseases. J. Cell. Biochem. 2019, 120, 8908–8918. [Google Scholar] [CrossRef] [PubMed]
  155. Asadi, M.R.; Hassani, M.; Kiani, S.; Sabaie, H.; Moslehian, M.S.; Kazemi, M.; Ghafouri-Fard, S.; Taheri, M.; Rezazadeh, M. The Perspective of Dysregulated LncRNAs in Alzheimer’s Disease: A Systematic Scoping Review. Front. Aging Neurosci. 2021, 13, 709568. [Google Scholar] [CrossRef] [PubMed]
  156. Zhang, J.; Wang, R. Deregulated lncRNA MAGI2-AS3 in Alzheimer’s disease attenuates amyloid-beta induced neurotoxicity and neuroinflammation by sponging miR-374b-5p. Exp. Gerontol. 2021, 144, 111180. [Google Scholar] [CrossRef]
  157. Ding, Y.; Luan, W.; Wang, Z.; Cao, Y. LncRNA BDNF-AS as ceRNA regulates the miR-9-5p/BACE1 pathway affecting neurotoxicity in Alzheimer’s disease. Arch. Gerontol. Geriatr. 2022, 99, 104614. [Google Scholar] [CrossRef] [PubMed]
  158. He, L.; Chen, Z.; Wang, J.; Feng, H. Expression relationship and significance of NEAT1 and miR-27a-3p in serum and cerebrospinal fluid of patients with Alzheimer’s disease. BMC Neurol. 2022, 22, 203. [Google Scholar] [CrossRef] [PubMed]
  159. Zhao, M.Y.; Wang, G.Q.; Wang, N.N.; Yu, Q.Y.; Liu, R.L.; Shi, W.Q. The long-non-coding RNA NEAT1 is a novel target for Alzheimer’s disease progression via miR-124/BACE1 axis. Neurol. Res. 2019, 41, 489–497. [Google Scholar] [CrossRef] [PubMed]
  160. Riku, Y.; Seilhean, D.; Duyckaerts, C.; Boluda, S.; Iguchi, Y.; Ishigaki, S.; Iwasaki, Y.; Yoshida, M.; Sobue, G.; Katsuno, M. Pathway from TDP-43-Related Pathology to Neuronal Dysfunction in Amyotrophic Lateral Sclerosis and Frontotemporal Lobar Degeneration. Int. J. Mol. Sci. 2021, 22, 3843. [Google Scholar] [CrossRef] [PubMed]
  161. Gumina, V.; Onesto, E.; Colombrita, C.; Maraschi, A.; Silani, V.; Ratti, A. Inter-Species Differences in Regulation of the Progranulin-Sortilin Axis in TDP-43 Cell Models of Neurodegeneration. Int. J. Mol. Sci. 2019, 20, 5866. [Google Scholar] [CrossRef] [PubMed]
  162. Tollervey, J.R.; Curk, T.; Rogelj, B.; Briese, M.; Cereda, M.; Kayikci, M.; König, J.; Hortobágyi, T.; Nishimura, A.L.; Župunski, V.; et al. Characterising the RNA targets and position-dependent splicing regulation by TDP-43; implications for neurodegenerative diseases. Nat. Neurosci. 2011, 14, 452–458. [Google Scholar] [CrossRef] [PubMed]
  163. Fan, Y.; Xiao, B.; Zhang, M. Potential Intersections between lncRNA, Vascular Cognitive Impairment, and Immunization Strategies: Insights and Future Directions. Vaccines 2024, 12, 251. [Google Scholar] [CrossRef] [PubMed]
  164. Qi, D.; Hou, X.; Jin, C.; Chen, X.; Pan, C.; Fu, H.; Song, L.; Xue, J. HNSC exosome-derived MIAT improves cognitive disorders in rats with vascular dementia via the miR-34b-5p/CALB1 axis. Am. J. Transl. Res. 2021, 13, 10075–10093. [Google Scholar] [PubMed]
  165. Wang, P.; Mao, S.; Yi, T.; Wang, L. LncRNA MALAT1 Targets miR-9-3p to Upregulate SAP97 in the Hippocampus of Mice with Vascular Dementia. Biochem. Genet. 2023, 61, 916–930. [Google Scholar] [CrossRef] [PubMed]
  166. Vijayan, M.; Reddy, P.H. Non-Coding RNAs Based Molecular Links in Type 2 Diabetes, Ischemic Stroke, and Vascular Dementia. J. Alzheimer’s Dis. 2020, 75, 353–383. [Google Scholar] [CrossRef] [PubMed]
  167. Firat, H.; Nazi, S.A.A.; Bier, J.C.; Blanc, F.; Boutillier, S.; Danilin, S.; David, R.; Démonet, J.F.; Dubois, B.; Frisoni, G.B.; et al. ADDIA Consortium; ADKIT Consortium. lncRNAs as a novel source of diagnostic applications for early Alzheimer’s disease and other dementia types. Alzheimer’s Dement. 2020, 16, e039788. [Google Scholar] [CrossRef]
  168. Chen, L.; Shan, G. CircRNA in cancer: Fundamental mechanism and clinical potential. Cancer Lett. 2021, 505, 49–57. [Google Scholar] [CrossRef] [PubMed]
  169. Qin, T.; Li, J.; Zhang, K.Q. Structure, regulation, and function of linear and circular long non-coding RNAs. Front. Genet. 2020, 11, 150. [Google Scholar] [CrossRef]
  170. Lu, M. Circular RNA: Functions, applications and prospects. ExRNA 2020, 2, 1. [Google Scholar] [CrossRef]
  171. Puri, S.; Hu, J.; Sun, Z.; Lin, M.; Stein, T.D.; Farrer, L.A.; Wolozin, B.; Zhang, X. Identification of circRNAs linked to Alzheimer’s disease and related dementias. Alzheimer’s Dement. 2023, 19, 3389–3405. [Google Scholar] [CrossRef] [PubMed]
  172. Han, B.; Chao, J.; Yao, H. Circular RNA and its mechanisms in disease: From the bench to the clinic. Pharmacol. Therapeut. 2018, 187, 31–44. [Google Scholar] [CrossRef] [PubMed]
  173. Zhang, Y.; Yu, F.; Bao, S.; Sun, J. Systematic Characterization of Circular RNA-Associated CeRNA Network Identified Novel circRNA Biomarkers in Alzheimer’s Disease. Front. Bioeng. Biotechnol. 2019, 7, 222. [Google Scholar] [CrossRef] [PubMed]
  174. Yang, H.; Wang, H.; Shang, H.; Chen, X.; Yang, S.; Qu, Y.; Ding, J.; Li, X. Circular RNA circ_0000950 promotes neuron apoptosis, suppresses neurite outgrowth and elevates inflammatory cytokines levels via directly sponging miR-103 in Alzheimer’s disease. Cell Cycle 2019, 18, 2197–2214. [Google Scholar] [CrossRef] [PubMed]
  175. Lu, Y.; Tan, L.; Wang, X. Circular HDAC9/microRNA-138/Sirtuin-1 Pathway Mediates Synaptic and Amyloid Precursor Protein Processing Deficits in Alzheimer’s Disease. Neurosci. Bull. 2019, 35, 877–888. [Google Scholar] [CrossRef] [PubMed]
  176. Zhao, Y.; Alexandrov, P.N.; Jaber, V.; Lukiw, W.J. Deficiency in the Ubiquitin Conjugating Enzyme UBE2A in Alzheimer’s Disease (AD) is Linked to Deficits in a Natural Circular miRNA-7 Sponge. Genes 2016, 7, 116. [Google Scholar] [CrossRef] [PubMed]
  177. Xu, X.; Gu, D.; Xu, B.; Yang, C.; Wang, L. Circular RNA circ_0005835 promotes promoted neural stem cells proliferation and differentiate to neuron and inhibits inflammatory cytokines levels through miR-576-3p in Alzheimer’s disease. Environ. Sci. Pollut. R. 2022, 29, 35934–35943. [Google Scholar] [CrossRef] [PubMed]
  178. Zhang, Q.; Chen, B.; Yang, P.; Wu, J.; Pang, X.; Pang, C. Bioinformatics-based study reveals that AP2M1 is regulated by the circRNA-miRNA-mRNA interaction network and affects Alzheimer’s disease. Front. Genet. 2022, 13, 1049786. [Google Scholar] [CrossRef] [PubMed]
  179. Liu, L.; Chen, X.; Chen, Y.H.; Zhang, K. Identification of Circular RNA hsa_Circ_0003391 in Peripheral Blood Is Potentially Associated With Alzheimer’s Disease. Front. Aging Neurosci. 2020, 12, 601965. [Google Scholar] [CrossRef] [PubMed]
  180. Li, Y.; Han, X.; Fan, H.; Sun, J.; Ni, M.; Zhang, L.; Fang, F.; Zhang, W.; Ma, P. Circular RNA AXL increases neuron injury and inflammation through targeting microRNA-328 mediated BACE1 in Alzheimer’s disease. Neurosci. Lett. 2022, 776, 136531. [Google Scholar] [CrossRef] [PubMed]
  181. Huang, Y.; Liao, X.; Luo, J.; Liu, H.; Zhong, S.; Chen, J. Expression of circular RNAs in the vascular dementia rats. Neurosci. Lett. 2020, 735, 135087. [Google Scholar] [CrossRef] [PubMed]
  182. Li, L.; Xu, Y.; Zhao, M.; Gao, Z. Neuro-protective roles of long non-coding RNA MALAT1 in Alzheimer’s disease with the involvement of the microRNA-30b/CNR1 network and the following PI3K/AKT activation. Exp. Mol. Pathol. 2020, 117, 104545. [Google Scholar] [CrossRef] [PubMed]
  183. Zhuang, J.; Cai, P.; Chen, Z.; Yang, Q.; Chen, X.; Wang, X.; Zhuang, X. Long noncoding RNA MALAT1 and its target microRNA-125b are potential biomarkers for Alzheimer’s disease management via interactions with FOXQ1, PTGS2 and CDK5. Am. J. Transl. Res. 2020, 12, 5940–5954. [Google Scholar] [PubMed]
  184. Zhou, B.; Li, L.; Qiu, X.I.N.; Wu, J.; Xu, L.E.I.; Shao, W. Long non-coding RNA ANRIL knockdown suppresses apoptosis and pro-inflammatory cytokines while enhancing neurite outgrowth via binding microRNA-125a in a cellular model of Alzheimer’s disease. Mol. Med. Rep. 2020, 22, 1489–1497. [Google Scholar] [CrossRef] [PubMed]
  185. Zhang, Y.Y.; Bao, H.L.; Dong, L.X.; Liu, Y.; Zhang, G.W.; An, F.M. Silenced lncRNA H19 and up-regulated microRNA-129 accelerates viability and restrains apoptosis of PC12 cells induced by Aβ25-35 in a cellular model of Alzheimer’s disease. Cell Cycle 2021, 20, 112–125. [Google Scholar] [CrossRef] [PubMed]
  186. Zhou, Y.; Ge, Y.; Liu, Q.; Li, Y.X.; Chao, X.; Guan, J.J.; Diwu, Y.C.; Zhang, Q. LncRNA BACE1-AS Promotes Autophagy-Mediated Neuronal Damage Through The miR-214-3p/ATG5 Signalling Axis In Alzheimer’s Disease. Neuroscience 2021, 455, 52–64. [Google Scholar] [CrossRef] [PubMed]
  187. Zhou, L.; Huang, X.; Li, H.; Wang, J.; Lu, Z. Triptolide improves Alzheimer’s disease by regulating the NF-κB signaling pathway through the lncRNA NEAT1microRNA 361-3pTRAF2 axis. Exp. Ther. Med. 2023, 26, 440. [Google Scholar] [PubMed]
  188. Zhang, N.; Gao, Y.; Yu, S.; Sun, X.; Shen, K. Berberine attenuates Aβ42-induced neuronal damage through regulating circHDAC9/miR-142-5p axis in human neuronal cells. Life Sci. 2020, 252, 117637. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The mechanisms of dementia pathogenesis through ceRNA interactions. (a) Increased expression of lncRNA or circRNA acts as a miRNA sponge, inhibiting miRNA expression. It leads to the high expression of a target gene involved in dementia. (b) Downregulated expression of lncRNA or circRNA induces upregulation of miRNA expression, resulting in decreased mRNA expression levels of a target gene associated with dementia. MRE: miRNA response elements.
Figure 1. The mechanisms of dementia pathogenesis through ceRNA interactions. (a) Increased expression of lncRNA or circRNA acts as a miRNA sponge, inhibiting miRNA expression. It leads to the high expression of a target gene involved in dementia. (b) Downregulated expression of lncRNA or circRNA induces upregulation of miRNA expression, resulting in decreased mRNA expression levels of a target gene associated with dementia. MRE: miRNA response elements.
Ijms 25 06190 g001
Figure 2. The complex interactions observed among miRNAs (light blue rectangles), lncRNAs (green rhombuses), and target genes (yellow ovals) in the context of AD with 10 interactions denoted by a blue dotted box, FTD with 1 interaction marked by a purple dotted box, and VD with 2 interactions identified within an orange dotted box. The interactions reflect the regulatory roles of miRNAs in inhibiting target genes, while lncRNAs act as miRNA sponges, thereby suppressing the inhibitory activity of miRNAs and contributing to the intricate network of gene expression regulation in dementia-related disorders.
Figure 2. The complex interactions observed among miRNAs (light blue rectangles), lncRNAs (green rhombuses), and target genes (yellow ovals) in the context of AD with 10 interactions denoted by a blue dotted box, FTD with 1 interaction marked by a purple dotted box, and VD with 2 interactions identified within an orange dotted box. The interactions reflect the regulatory roles of miRNAs in inhibiting target genes, while lncRNAs act as miRNA sponges, thereby suppressing the inhibitory activity of miRNAs and contributing to the intricate network of gene expression regulation in dementia-related disorders.
Ijms 25 06190 g002
Figure 3. The intricate interactions among miRNAs (light blue rectangles), circRNAs (green rhombuses), and target genes (yellow ovals) in AD (blue dotted box) with 10 interactions. The figure demonstrates miRNAs’ regulatory roles in inhibiting target genes, alongside circRNAs functioning as miRNA sponges to suppress miRNA activity, thus forming a complex gene expression network in AD.
Figure 3. The intricate interactions among miRNAs (light blue rectangles), circRNAs (green rhombuses), and target genes (yellow ovals) in AD (blue dotted box) with 10 interactions. The figure demonstrates miRNAs’ regulatory roles in inhibiting target genes, alongside circRNAs functioning as miRNA sponges to suppress miRNA activity, thus forming a complex gene expression network in AD.
Ijms 25 06190 g003
Figure 4. The key molecules in dementia-related diseases. (a) AD primarily results from the accumulation of Aβ in cells due to increased expression of BACE1. Although miRNAs can complementarily bind to BACE1 mRNA, lncRNAs and circRNAs may act as miRNA sponges instead. (b) FTD arises from mislocalization of TDP-43, normally located in the nucleus, leading to increased expression in the cytoplasm. This cytoplasmic TDP-43 suppresses GRN expression, potentially regulated by downregulated miRNAs and lncRNAs. (c) LBD is associated with Lewy bodies formed by misfolded α-synuclein. (d) The specific genetic factors involved in VD, potentially due to vascular damage, are not precisely known, although multiple genetic factors are suggested to be involved. AD: Alzheimer’s disease; APP: Amyloid-beta precursor protein; BACE1: beta-Secretase; MAGI2-AS3: Membrane Associated Guanylate Kinase, WW, and PDZ Domain Containing 2 Antisense RNA 3; BDNF-AS: Brain-Derived Neurotrophic Factor-AS; NEAT1: Nuclear-Enriched Abundant Transcript; FTD: frontotemporal dementia; GRN: granulin precursor; PGRN: progranulin; MALAT1: Metastasis Associated Lung Adenocarcinoma Transcript 1; TDP-43: Transactivation response DNA binding protein 43 kDa; LBD: Lewy body dementia; SNCA: alpha-synuclein; VD: vascular dementia.
Figure 4. The key molecules in dementia-related diseases. (a) AD primarily results from the accumulation of Aβ in cells due to increased expression of BACE1. Although miRNAs can complementarily bind to BACE1 mRNA, lncRNAs and circRNAs may act as miRNA sponges instead. (b) FTD arises from mislocalization of TDP-43, normally located in the nucleus, leading to increased expression in the cytoplasm. This cytoplasmic TDP-43 suppresses GRN expression, potentially regulated by downregulated miRNAs and lncRNAs. (c) LBD is associated with Lewy bodies formed by misfolded α-synuclein. (d) The specific genetic factors involved in VD, potentially due to vascular damage, are not precisely known, although multiple genetic factors are suggested to be involved. AD: Alzheimer’s disease; APP: Amyloid-beta precursor protein; BACE1: beta-Secretase; MAGI2-AS3: Membrane Associated Guanylate Kinase, WW, and PDZ Domain Containing 2 Antisense RNA 3; BDNF-AS: Brain-Derived Neurotrophic Factor-AS; NEAT1: Nuclear-Enriched Abundant Transcript; FTD: frontotemporal dementia; GRN: granulin precursor; PGRN: progranulin; MALAT1: Metastasis Associated Lung Adenocarcinoma Transcript 1; TDP-43: Transactivation response DNA binding protein 43 kDa; LBD: Lewy body dementia; SNCA: alpha-synuclein; VD: vascular dementia.
Ijms 25 06190 g004
Table 1. List of differentially expressed miRNAs related to AD.
Table 1. List of differentially expressed miRNAs related to AD.
miRNA Expression in ADmiRNATarget GeneClinical ValueFunctionStudy ModelReference
UPmiR-20b-5pAPPN/AReduce intracellular Ca2+ transients during neuronal membrane depolarizationHuman tissues and in vitro[57]
miR-140PINK1Biomarker and therapeutic targetImprove amyloid pathology and mitochondrial dysfunction while inhibiting cellular autophagyIn vivo[58]
miR-425-5pHSPB8Therapeutic targetReduce cell apoptosis and tau phosphorylationHuman tissues and in vitro[59]
miR-10b-5pHOXD10Therapeutic targetReduce nerve cell apoptosis, inflammatory response, and oxidative stressIn vivo[60]
miR-455-5pCPEB1Therapeutic targetSynaptic plasticity and memory disordersIn vivo[61]
miR-592KIAA0319Therapeutic targetPromotion of oxidative stress injury in astrocytesIn vivo and in vitro[62]
miR-1273g-3pTIMM13Biomarker and therapeutic targetMitochondrial dysfunctionHuman tissues and in vitro[63]
miR-485-3pCD36Biomarker and therapeutic targetInhibit microglial Aβ phagocytosisIn vivo and in vitro[64]
miR-134-5pCREB-1Therapeutic targetRegulation of long-term plasticity and cellular correlationIn vivo and in vitro[65]
BDNF
miR-384-Diagnostic biomarkerInvolved in immune responseHuman samples[66]
miR-1229-3pSORL1Therapeutic targetEngaged in the processing and movement of AβIn vitro[67]
miR-29a-3pC1QTNF6Diagnostic biomarkerMigration of neurons and evolution of the nervous systemHuman samples[68]
ROBO1
DAAM2
let-7i-5p-Diagnostic biomarkerRegulating APP and BACE1, leading to AD pathologyHuman samples[69]
miR-15a-5p
miR-34aVAMP2Therapeutic targetAbnormalities in energy metabolism, resting state network activity, and synaptic plasticityHuman tissues and in vivo[70]
SYT1
DOWNmiR-92a-3pSYNJ1Diagnostic biomarkerConnected to protein and lipid pathways, transcription, structural function, and amyloid-beta clearance/cell signalingHuman samples[68]
CBLN4
BCL2L2
NEFH
REST
miR-132FOXA1Therapeutic targetImprove cognitive impairmentIn vivo[71]
miR-326VAV1Therapeutic targetTau phosphorylation and neuronal apoptosisIn vivo[72]
miR-146a-5pNkd2Therapeutic targetInhibit LPS/Aβ-induced neuroinflammation and regulate a microglial phenotypeIn vivo and in vitro[73]
miR-195ApoE4Therapeutic targetRegulate tau hyperphosphorylation and secretionHuman tissues and in vivo[74]
miR-103BACE1Prognosis biomarkerEnhance neurite outgrowth and reduce neuronal apoptosisHuman tissues[75]
miR-107
miR-455-3p-N/AImprove neuronal activity and overall brain functionIn vivo[76]
miR-146aTRAF6N/ASuppress astrocyte inflammation in ADIn vivo[77]
miR-149BACE1Diagnostic biomarkerReduce accumulation of Aβ and improve the viability of neuronsHuman serum and in vitro[78]
miR-9-5pBACE1Diagnostic biomarkerRegulate differentiation of post-mitotic neurons from neural progenitor cellsHuman serum[79]
SIRT1
miR-107FGF7N/AAmeliorate Aβ-induced inflammation and apoptosisHuman serum and in vitro[80]
miR-23bGnT-IIITherapeutic targetInhibit oxidative stress and activate the Akt/GSK-3β signaling pathwayIn vivo and in vitro[81]
miR-29c-3pBACE1Therapeutic targetAβ-induced suppression of neuronal viability and increase in apoptosisIn vivo[82]
miR-31APPTherapeutic targetImprove cognitive function and prevent the progression of the diseaseIn vivo and in vitro[83]
BACE1
miR-212PDCD4N/AMitigate Aβ25-35 induced neurotoxicity through modulation of the PI3K/AKT pathwayHuman serum and in vitro[84]
miR-22-3pSOX9Therapeutic targetEnhance apoptosis attenuation and reduce Aβ accumulationIn vivo and in vitro[85]
miR-126a-3pEFHD2Therapeutic targetConsolidate contextual fear memoryIn vivo and in vitro[86]
miR-22GSDMDTherapeutic targetEnhancing memory and motor abilitiesIn vivo[87]
miR-92a-3pMAPTDiagnostic biomarkerControl the expression of tau in a neuroblastomaHuman samples and in vitro[88]
miR-320a
miR-132MAPTTherapeutic targetPartially recovered tau metabolism and memoryHuman tissues, in vivo and in vitro[89]
miR-212
miR-188-3pBACE1Therapeutic targetEnhanced synaptic and cognitive function through decreased neuroinflammation and Aβ due to MAGL inhibitionIn vivo and in vitro[90]
miR-132-3pFOXO1aN/AHyperphosphorylation of tauHuman tissues[91]
miR-512cFLIPN/AApoptosis initiating factor, APAF-1 activity, activated caspase-3, elevated caspase-4 and caspase-8, and the TUNEL assay was negative in the regions where neurons displayed hyperphosphorylated tauHuman samples[92]
MCL1
N/A: not available.
Table 2. List of differentially expressed miRNAs involved in other dementia-related diseases.
Table 2. List of differentially expressed miRNAs involved in other dementia-related diseases.
Name of DiseasemiRNA Expression in DiseasemiRNATarget GeneClinical ValueFunctionStudy ModelReference
FTDUPmiR-29bGRNTherapeutic targetInduce progranulin deficiency and development of neurodegenerative diseasesIn vitro[110]
miR-659-3pGRNN/ANeurotrophic, anti-inflammatory activity and act as neuroprotective against oxygen/glucose deprivation, oxidative injury, and hypoxia stressIn vivo and in vitro[111]
DOWNmiR-632GRNDiagnostic biomarkerPrevents apoptosis, averting degenerative changes in the frontal and temporal lobesHuman samples[112]
miR-132TMEM106BTherapeutic targetDisrupt endosomal-lysosomal pathways, trapping PGRN in TMEM106B-positive compartments, elevating intracellular PGRN levelsHuman tissues and in vitro[113]
miR-212
miR-124CHMP2BTherapeutic targetReduce AMPAR levels and partial rescue of behavioral deficitsHuman tissues and in vivo[114]
miR-127-3p-Diagnostic biomarkerRegulate neuronal differentiationHuman samples[115]
LBDUPmiR-7SNCAN/AThe process of forming dopamine-producing neuronsHuman tissues and in vitro[116]
miR-153
VDUPmiR-210-5pSnap25Therapeutic targetSynaptic loss impacting spatial learning and memoryIn vivo[117]
miR-134-5pFoxp2Therapeutic targetContribute to speech and language and associated neurodevelopmental disordersIn vivo and in vitro[118]
miR-93TSPAN5Therapeutic targetReduce inflammation and promote positive regulation of the TLR4 signaling pathwayIn vivo[119]
miR-150HOXA1Biomarker and therapeutic targetIncrease cell apoptosisIn vivo and in vitro[120]
miR-134Cofilin 2N/ARegulate oxidative stress and autophagyIn vivo[121]
miR-154-5pPRKAA2Biomarker and therapeutic targetImpair EPC function and angiogenesisHuman samples and in vivo[122]
miR-181aPINK1N/AAlleviate mitochondrial dysfunction and improve cognitive functionIn vivo[123]
Parkin
DOWNmiR-216aRSK2Therapeutic targetRegulate oxidative stress and neuroinflammationIn vivo[124]
miR-132-3pRASA1Therapeutic targetImprove neuronal and synaptic dysfunction by activating the Ras/Akt/GSK-3β pathwayIn vivo[125]
miR-322-5pTSPAN5Therapeutic targetImprove cell apoptosis, inflammatory response, and cognitive functionIn vivo and in vitro[126]
N/A: not available.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Kim, J.-m.; Kim, W.R.; Park, E.G.; Lee, D.H.; Lee, Y.J.; Shin, H.J.; Jeong, H.-s.; Roh, H.-Y.; Kim, H.-S. Exploring the Regulatory Landscape of Dementia: Insights from Non-Coding RNAs. Int. J. Mol. Sci. 2024, 25, 6190. https://doi.org/10.3390/ijms25116190

AMA Style

Kim J-m, Kim WR, Park EG, Lee DH, Lee YJ, Shin HJ, Jeong H-s, Roh H-Y, Kim H-S. Exploring the Regulatory Landscape of Dementia: Insights from Non-Coding RNAs. International Journal of Molecular Sciences. 2024; 25(11):6190. https://doi.org/10.3390/ijms25116190

Chicago/Turabian Style

Kim, Jung-min, Woo Ryung Kim, Eun Gyung Park, Du Hyeong Lee, Yun Ju Lee, Hae Jin Shin, Hyeon-su Jeong, Hyun-Young Roh, and Heui-Soo Kim. 2024. "Exploring the Regulatory Landscape of Dementia: Insights from Non-Coding RNAs" International Journal of Molecular Sciences 25, no. 11: 6190. https://doi.org/10.3390/ijms25116190

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