Abstract
MicroRNAs are small non-coding RNAs evolutionary conserved molecules. They regulate cellular processes, including RNA silencing, post-translational gene expression and neurodegeneration. MicroRNAs are involved with human diseases such as cancer, Alzheimer’s disease (AD) and others. Interestingly, cerebrospinal fluids (CSF) and the blood of AD patients have altered expressions of many RNAs, which may serve as potential peripheral biomarkers. The intensive investigation from our lab revealed that MicroRNA-455-3p (miR-455-3p) is a strong candidate as a potential biomarker and therapeutic target for AD. Several genes implicated in the pathogenesis of AD are directly targeted by miR-455-3p. Several years of our lab research revealed that miR-455-3p regulates important physiological processes associated with AD, such as the processing of the amyloid precursor protein (APP), TGF-β signaling, the regulation of oxidative stress, mitochondrial biogenesis, and synaptic damages. The expression of miR-455-3p in mild cognitive impaired subjects and AD patients pointed out its involvement in AD progression. Recently, our lab generated both transgenic and knockout mice for miR-455-3p. Interestingly miR-455-3p transgenic mice showed superior cognitive learning, improved memory and extended lifespan compared to age matched wild-type mice, whereas mir-455-3-p knockout mice showed cognitive decline and reduced lifespan. Information derived from mouse models further demonstrated the advantageous impact of miR-455-3p on dendritic growth, synaptogenesis, and mitochondrial biogenesis in preventing the onset and progression of AD. The identification of miR-455-3p as a biomarker was suggested by its presence in postmortem AD brains, B-lymphocytes, and fibroblasts. Our hypothesis that miR-455-3p could be a peripheral biomarker and therapeutic target for AD.
Keywords: Alzheimer’s Disease, amyloid precursor protein, Biomarker, Cognitive behavior, microRNA-455-3-p, mitochondria, synaptogenesis
Introduction
The expression of genetic information involves transcription of ribonucleic acid (RNA) molecules from deoxyribonucleic acid (DNA) and their role in protein biosynthesis. Mature messenger RNAs are linear entities with 5′ and 3′ termini that reflect start and stop of the RNA polymerase on the DNA template (Jacob and Monod, 1961). Transcription process of the whole eukaryotic genome also produces a plethora of non-protein-coding RNA species with complicated overlapping patterns of expression and control. Later numerous regulatory RNAs of all shapes and sizes have been discovered (Amaral et al., 2008; Ponting et al., 2009). Noncoding transcripts are classified as either housekeeping or regulatory. Housekeeping noncoding RNAs (ncRNA), including as ribosomal, transfer, small nuclear, and small nucleolar RNAs. Short regulatory noncoding RNAs include microRNAs (miRNA), small interfering RNAs, and Piwi-associated RNAs (Carthew and Sontheimer, 2009; Malone and Hannon, 2009). In most instances, ncRNAs are directly involved in the regulation of most cell processes, including epigenetic control, gene transcription, translation, RNA turnover, chromosomal organization, and genome defense, with key roles in cellular developmental and proliferation programs (Cech and Steitz, 2014; Huntzinger and Izaurralde, 2011; Morris and Mattick, 2014; Rinn and Chang, 2012; Sabin et al., 2013).
miRNA molecules have emerged as key regulators in the expression and function of eukaryotic genomes over the last 2 decades. This regulation, mostly inhibitory in nature, can occur at the most fundamental levels of genome function, such as chromatin structure, chromosome segregation, transcription, RNA processing, RNA stability, and translation. miRNAs inhibit the expression of mRNA targets by promoting translational repression, and mRNA degradation. Translational repression is thought to occur in four different ways: inhibition of translation initiation, the suppression of translation elongation, co-translational protein degradation, and premature termination of translation (Eulalio et al., 2008; Wu and Belasco, 2008). In animals, target degradation begins with rapid deadenylation, which is typically followed by decapping and subsequent degradation of the mRNA, but in plants, target degradation begins with endonucleolytic cleavage catalyzed by the argonaute proteins (Krol et al., 2010; Voinnet, 2009). A mature miRNA typically attaches to the 3′ untranslated region (UTR) of the designated messenger RNA (mRNA) as a part of the RNA-induced silencing complex (RISC) and inhibit the translation process (Figure. 1) (Iwakawa and Tomari, 2015). The fundamental determinant of target recognition is a brief sequence complementarity between the miRNA seed sequence (nucleotides 2–7) and the targeted mRNA (Lewis et al., 2005).
Figure 1:

Overview of miRNA biogenesis. miRNAs are transcribed by RNA polymerases II or III as pri-mRNA. primary miRNAs (pri-miRNAs) are recognized and excised by the microprocessor complex, composed by DiGeorge Syndrome Critical Region 8 (DGCR8) and Drosha, into a hairpin-shaped precursor-miRNAs (pre-miRNAs) via the canonical pathway in the nucleus. The pre-miRNA is exported to the cytoplasm in an Exportin 5/RanGTP-dependent manner. In the cytoplasm, dicer cleaves pre-miRNAs into tiny double-stranded miRNAs. One of the strands of the miRNA duplex is incorporated into the Argonaute (AGO) family of proteins to form a miRNA-induced silencing complex (miRISC). The miRISC complex then mediates the recognition of the target mRNA at the 3′ untranslated region, which results in mRNA instability or translational repression. The gene regulatory power of cytoplasmic miRISC typically culminates via induction of translation inhibition and mRNA poly(A) deadenylation.
Depending on their origin and expression patterns, certain miRNAs are highly tissue specific and restricted to specific cellular niches, whereas other miRNA families are expressed throughout the human body (Ambros, 2004; Bartel, 2004; Ludwig et al., 2016). In several occasions, miRNAs are encapsulated in a variety of structures, including exosomes, microvesicles, and apoptotic bodies, which release them into the extracellular space to regulate intracellular communication (Kosaka et al., 2013; Turchinovich et al., 2013). Recently scientists are considering circulating miRNA as potential biomarkers for cancer, ageing and neurological disorders (Kosaka et al., 2010; Kumar et al., 2017a; Kumar et al., 2017b; Olivieri et al., 2017).
Alzheimer’s disease (AD) is a neurological disorder that progresses over time and is heterogeneous. The hallmark of AD is memory loss, cognitive function impairment, and changes in behavioral and personality traits (LaFerla et al., 2007; Mattson, 2004; Reddy et al., 2010, Oliver and Reddy, 2019; Morton et al., 2021). Early-onset familial AD caused mostly by genetic mutations while late-onset sporadic AD is induced by ageing and other lifestyle factors (Amakiri et al., 2019; Sehar et al., 2022; Rawat et al., 2022). Mutations in amyloid precursor protein (APP), presenilin 1 (PS1), presenilin 2 (PS2), apolipoprotein E4 (APOE4), srotilin related receptor 1, clusterin, complement component receptor 1, CD2 Associated Protein (CD2AP), Aph-1 Homolog A (APHA1), and membrane-spanning 4-domains subfamily A (MS4A4/MS4A6E) genes have been reported in different AD patients (Mao and Reddy, 2011).
AD is marked by accumulation of intracellular neurofibrillary tangles (NFTs) and extracellular amyloid-β (Aβ) plaques in brain leading to synaptic damage, proliferation of reactive astrocytes and activated microglia, an age-dependent imbalance in hormones, and structural and functional changes in mitochondria (DeKosky et al., 1996; Du et al., 2010; Reddy, 2006; Reddy and Beal, 2008; Reddy et al., 2012; Swerdlow, 2011; Tampellini and Gouras, 2010; Terry et al., 1991). Although a great deal of work has been done in understanding the pathophysiology of AD, there are currently no identifiable biomarkers, medications, or other interventions that can stop AD or decrease its progression. The hunt for novel, non-invasive, peripherally available, early detectable biomarkers associated with AD and diseases related to Alzheimer’s disease (ADRD) is still progressing. Though many miRNAs that regulate AD genes to regulate AD pathogenesis and are involved in AD-related pathways have been found (Chopra et al., 2021; Kim et al., 2015; Kumar and Reddy, 2016; Long and Lahiri, 2011; Long et al., 2019; Ma et al., 2017; Vilardo et al., 2010), none of them can be definitively identified as an established biomarker for AD detection.
We highlight the relevance of miR-455-3p in AD in this review, mostly based on our lab’s (Reddy lab) findings, which are corroborated by other researchers. These findings lead us to establish miR-455-3p as a peripheral biomarker that can be found in the blood, CSF, fibroblast cells, and postmortem brains of AD patients.
1.1. Expression and biogenesis of miRNA-455-3P
MiRNA-455, with a highly conserved expression pattern, is present as non-coding RNA in most of the species from Chordata to Mammalia (Kumar and Reddy, 2018a). MiRNA-455 precursor sequence is transcribed by the intron 10 of human COL27A1 gene (collagen type XXVII alpha 1 chain). This precursor has 96 base pairs and is originally located at human chromosome 9 at locus 9q32 (Kumar and Reddy, 2018a). Human miR-455-3p has two different isoforms: miR-455-3p and miR-455-5p. miR-455 has key roles in different types of cancer ; human non-small cell lung cancer (Gao et al., 2018), oral squamous cancer cells (Cheng et al., 2016), renal cancer (Yamada et al., 2018), skin cancer (Shoshan et al., 2015), colorectal cancer (Zheng et al., 2016), hepatocellular carcinoma (Qin et al., 2016), breast cancer (Li et al., 2017b), pancreatic cancer (Zhan et al., 2018), and prostate cancer (Arai et al., 2019). In this review, we focus on its role in AD.
For more than eight years, the Reddy laboratory has been investigating miRNAs in ageing and age-related neurodegenerative disease such as AD. Earlier we reviewed the potential of circulating miRNAs as a peripheral biomarker for AD and their biological relevance in AD diagnosis, progression, and therapeutics. We found several probable miRNA candidates in AD (Kumar and Reddy, 2016). miR-455-3p got our attention when we found high expression in a global microarray analysis of serum samples collected from AD patients (Kumar et al., 2017b). Another research group also reported upregulation of miR-455-3p in hippocampus of AD patients compared to healthy controls (Lau et al., 2013). Further, our lab verified and validated the level of miR-455-3p in different kinds of AD samples (Kumar and Reddy, 2018b). We reported an elevation of MIRNA 455-3-p level on AD postmortem brains, AD fibroblasts, AD B-lymphocytes, AD cell lines, and APP transgenic mice [40, 41] compared to the samples from healthy controls. Moreover, we investigated the prospective role of miR-455-3p in AD biogenesis in in-silico, in vitro and in vivo models.
Here, we shed light to the significance of our findings and connection of miR-455-3p in the pathology, progression and modulation of MCI and AD. First, we discuss possible molecular and cellular events where miR-455-3p is offering a protective role in AD pathogenesis. In the later sections, we discuss the superior features of miR-455-3p in mice models in terms of ageing and AD development. Considering all these findings, we emphasize that miR-455-3p as a solid peripheral biomarker to diagnose and identify AD at an early state.
1.2. Role of miRNA 455-3P in AD related molecular and cellular events
Key cellular features of AD include not only accumulation of NFTs and Aβ plaques but also proliferation of reactive astrocytes, loss of cholinergic neurons due to synaptic damage, loss of synaptic proteins, defective autophagy/mitophagy and structural and functional alteration of mitochondria. According to our lab findings and other scientific reports miR-455-3p is directly regulating to some of the above features while indirectly affecting the others (Kumar et al., 2021).
a. Amyloid precursor protein processing (APP) and Tau as targets for miRNA 455-3P
Our lab investigated the protective role of miR-455-3p against APP processing and Aβ formation (Kumar et al., 2019a). Initially, predictive consequential pairing of has-miR455-3p.1 and has-miR-455-3p.2 with complementary target genes was studied and the conserved miR-455-3p.2 binding site was found in 323 projected transcripts/human genes. Surprisingly, a good number of target genes are involved in AD pathogenesis such as APP, NGF, USP25, PDRG1, SMAD4, UBQLN1, SMAD2, TP73, VAMP2, HSPBAP1, and NRXN1 (Kumar and Reddy, 2018b). In-silico analysis was performed to understand the link of miR-455-3p with gene regulating AD pathogenesis (Kumar et al., 2019a). We analyzed miR-455-3p target genes utilizing various online miRNA bio-informatics algorithms; for example, diana-microt, microrna.org, mirdb, rna22-has, targetminer, and targetscan-vert. It showed two binding sites of miR-455-3p at 3′ UTR of the APP gene (ENST00000346798.3) at sequence positions 522–528 and 3139–3145. The miR-455-3p binding sequence at the 3’UTR of APP mRNA was similar in the human and mouse genome. It suggests that miR-455-3p has a critical role in APP gene regulation affecting the expression level of APP genes. To validate these results, a luciferase reporter assay was conducted on a vector having a wild-type APP 3’UTR sequence and on a control vector. Luciferase activity was significantly decreased in the cells transfected with miR-455-3p mimics compared to control. These results define that the 3’UTR site of mRNA from the APP protein contains conserved binding sites for miR-455-3p (Figure 2). Moreover, in our qRT-PCR analysis, mRNA levels of APP gene were significantly reduced in the cells transfected with miR-455-3p mimics while strikingly upregulated in the cells transfected with the miR-455-3p inhibitors (Kumar et al., 2019a).
Figure 2:

miR455-3p has a critical role in APP gene regulation as miR-455-3p binds to two sequence locations in the 3′ UTR of the APP gene. It ultimately decreases the level of amyloid generation and accumulation in brain.
The protective role of miR-455-3p in AD was reveled via modulation of amyloid-β protein precursor (APP) and amyloid-β (Aβ) levels in mouse neuroblastoma cells. Overexpression of miR-455-3p construct reduces the expression of mutant APP cDNA and c-terminal fragments of APP (C99 and C83). Furthermore, miR-455-3p significantly reduced levels of Aβ (40) and (42) in mutant APP cells compared to control (Figure 2) (Kumar et al., 2019a) which ultimately explains the mechanistic role of miR-455-3p in AD progression.
Supporting our data, another article identified APP, BACE1 (Beta-site APP cleaving enzyme-1) and TAU as direct targets for miR-455-3p cause overexpression of miR-455-3p leading to a reduction in the expression of these genes in neuroblastoma cells (Swingler et al., 2022). In addition, they found an increase in the protein level of APP, BACE1 and TAU in the hippocampus of miR-455 null mice. Such findings reinforce the direct role of miR-455 in AD progression (Figure 2).
b. miR-455-3p and TGF-β signaling
In our lab miR-455-3p target genes were analyzed and surprisingly one TGFβ receptor, TGFBR3L, and two downstream regulators of TGFβ signaling pathway were identified namely SMAD2 and SMAD4 (Kumar and Reddy, 2018b). Supporting evidences were reported in other articles where they identified SMAD3 as a target gene of miR-455-3p (Liu et al., 2021) and showed that miR-455 downregulated TGFBR3 (Gharanei et al., 2020). In the TGF-β pathway, once bioavailable TGF-β comes into contact of the target cell, it binds with TGF-β type II receptors (TβRII) followed by a stable complex formation with TGF-β type I receptor (TβRI) (Massagué, 1998; Shi and Massagué, 2003). The activated ligand-receptor complex is then internalized via endocytosis (Chen, 2009). SMAD2 and SMAD3 are receptor-regulated effector proteins (R-Smads), which are phosphorylated by the activated TβRI, resulting in making a complex with SMAD4 (also known as co-SMAD) which ultimately activates the nuclear accumulation of SMAD complex (Massagué et al., 2005). Hence, by regulating the expression of SMAD2 and SMAD4 expression, miR 255-3-p might play a definitive role in TGF-β pathway in different aspects of cellular function (Figure 3).
Figure 3:

miR455-3p activates TGF beta signaling by regulating PAK2 expression,which ultimately ameliorates Alzheimer’s disease progression.
Several other reports have supported our claim that miR-455-3p has a critical role in TGF-β pathway. First, Swingler et al., found that miR-455-3p directly targets SMAD2, ACVR2B (Activin Receptor Type IIB) and CHRDL1 (Chordin-like 1) which could potentially modulate its functional impact on TGFβ signaling (Swingler et al., 2012). CHRDL1 gene encodes an antagonist of bone morphogenetic protein 4 (BMP4). As activin and BMP4 are essential component of TGFβ pathway, it indicates that miR-455-3p directly regulates this pathway in different probable routes in different cell functions. They also reported that the expression of miR-455-3p was regulated by TGFβ ligands during chondrogenesis. The upregulation of miR-455-3p were also reported in primary parenchymal lung fibroblasts after TGF-β1 stimulation (Ong et al., 2017). They also found NGF (nerve growth factor) and DLD (dihydrolipoamide dehydrogenase) genes as a target for miR-455-3p and these genes are involved in the TGF-β1-related biological processes and signaling pathway. On the contrary, downregulation of miR-455-3p upon TGF-β1 stimulation were reported in idiopathic pulmonary fibrosis (IPF) by another scientist group (Milosevic et al., 2012). This discrepancy could result from variations in the use of one donor versus several, culture techniques, and duration of TGF-β1 stimulation. Later, Zeng and colleagues found that TGF-β–induced protein ZEB1 (Zinc finger E-box-binding homeobox 1) is one of the target genes for miR-455-3p and through which it inhibited TGF-β signaling in a feedback loop mechanism (Zeng et al., 2019). Similar regulatory role of miR-455-3p via inhibiting ZEB1 expression was also reported during TGF-β1 induced signaling in endothelial to mesenchymal transition (Zhang et al., 2020). Another article suggested that miR-455-3p promoted the TGF/SMAD signaling pathway in chondrocytes. miR-455-3p inhibited the expression of P21-activated kinase 2 (PAK2) by directly targeting the 3′UTRs of PAK2 mRNA which ultimately promoted the R-SMAD activation (Figure 2) (Hu et al., 2019).
TGF-β1 signaling critically contributes to the proper functioning and protection of the brain in many ways. TGF-β1 shields neurons against harm caused by trophic factor deficiency, excitotoxins, hypoxia/ischemia, and Aβ aggregates (Caraci et al., 2008; Dhandapani et al., 2003). TGF-β1 also increases the expression of the anti-apoptotic proteins, Bcl-2 and Bcl-xl and maintains mitochondrial membrane potential (Prehn et al., 1996). On the contrary, reduced expression of TGFβRII receptor and impairments of TGF-β-activated SMAD signaling in AD brain were reported in several occasions (Chalmers and Love, 2007; Lee et al., 2006; Tesseur et al., 2006; Ueberham et al., 2006) suggesting dysfunction of TGF-β pathway is probably a causal factor in AD progression. Hallmarks of AD pathogenesis like NFT accumulation (Chalmers and Love, 2007; Lee et al., 2006) and Aβ deposition (Wang et al., 2010) disrupt the TGF/SMAD signaling pathway in AD brain. Taken together, further investigation is necessary to completely understand the fascinating part that TGF/SMAD signaling controlled by miR-455-3p plays in the development or prevention of AD.
c. Role of miR455-3p in alleviating oxidative stress and cell survival in apoptosis
Our preliminary miR-455-3p mimic transfection studies provide crucial hints regarding their defensive functions in cell proliferation and viability. In our lab, we found that neuroblastoma cells treated with miR-455-3p mimics showed extended cell proliferation compared to the controls whereas cells transfected with miR-455-3p inhibitors did not show regular proliferation (Kumar et al., 2019a). In addition, we conducted apoptosis analysis utilizing Annexin V-FITC reagent and found treatment with miR-455-3p significantly reduced apoptotic cell death and increased level of cell survival. Zhang and colleagues also found that miR-455-3p increased cell proliferation, promotes cell survival in MC3T3-E1 osteoblast cells (Zhang et al., 2018). miR-455-3p also promoted cell proliferation in pancreatic cancer cells (Zhan et al., 2018) and in breast cancer cell lines (Li et al., 2017a). Therefore, miR-455-3p may target different genes regulating cell proliferation and cell survival.
Several genes (e.g., BAX, BMF, and HIF1) mediating oxidative stress were marked as target genes for miR-455-3p in our lab data (Kumar et al., 2019a). Individuals with MCI, AD and AD related dementia exhibit a notable degree of oxidative damage to their brains, which is linked to the aberrant build-up of Aβ and the formation of NFTs (Christen, 2000; Huang et al., 2016). A number of articles proposed that miR-455-3p protects cells from oxidative stress. miR-455-3p shields osteoblasts from oxidative stress by targeting HDAC2 (histone deacetylases 2) to trigger nuclear factor erythroid 2-related factor 2 (Nrf2)/antioxidant response elements (AREs) signaling (Zhang et al., 2018). Moreover, Wang et al. reported that miR-455-3p played a protective role against high glucose-induced oxidative stress and cell injury by targeting Hmgb1 (high-mobility group box 1) gene (Wang et al., 2022). miR-455-3p also reduced apoptosis in chondrocyte through targeting PTEN (phosphatase and tensin homolog) gene (Wen et al., 2020) in PI3K/AKT pathway (Wen et al., 2020). In an oxygen and glucose deprivation (OGD) cell culture model, miR-455-3p alleviated oxidative stress related inflammatory responses by targeting SOX2-OT (sex-determining region Y-box 2 overlapping transcript) and TRAF6 (tumor necrosis factor receptor associated factor 6) genes (Gu et al., 2020). Strikingly, miR-455-3p protected neurons from oxidative injury by targeting small nucleolar RNA host gene 15 (SNHG15) and tumor protein p53 inducible nuclear protein 1 (TP53INP1) mRNAs (Fan et al., 2021). Taken together, we would like to suggest that miR-455-3p might have a distinctive role in mediating oxidative stress and inflammation in AD pathogenesis.
d. miR455-3p and mitochondrial biogenesis
Given the development of the AD are predominantly caused by structural and functional abnormalities in the mitochondria (Calkins and Reddy, 2011; Reddy, 2011), our lab evaluated the impact of miR-455-3p on the mRNA and protein levels of mitochondrial biogenesis genes (PGC1α, NRF1, TFAM). We found those were significantly increased in cells co-transfected with miR-455-3p and mutant APP relative to cells transfected with mutant APP alone. Further, we investigated the effect of miR-455-3p on mitochondrial dynamics genes. mRNA and protein levels of mitochondrial fission proteins (DRP1 and FIS1) were upregulated in the cells co-transfected with miR-455-3p and mutant APP cDNA. On the contrary, and fusion proteins (OPA1, Mfn1, and Mfn2) were decreased in the same experiment after miR-455-3p treatment. In addition, miR-455-3p also showed protective effects against mutant APP-induced mitochondrial fragmentations and repaired the abnormal mitochondrial dynamics, as captured in our electron microscopic analysis of neuroblastoma cells co-transfected with mutant APP cDNA and miR-455-3p (Kumar et al., 2019a). Supporting this, mitochondrial biogenesis and dynamics were improved in our miR-455-3p TG (transgenic) mice, while these were reduced in KO (knock out) mice what will be elaborated in later sections of this article. These findings lead us to the conclusion that miR-455-3p overexpression preserves normal mitochondrial activity by lessening the harmful effects of Aβ on mitochondria.
It is debated, nevertheless, how miR-455-3p controls mitochondrial activities. miR-455-3p targets hypoxia-inducible factor 1 α inhibitor (HIF1an) gene which has a key regulatory functions in mitochondrial biogenesis via interacting HIF1an- AMP-activated kinase α1 subunit (AMPK α1)-PGC1α signaling network (O’Hagan et al., 2009; Yun et al., 2019; Zhang et al., 2015). Through the inhibition of HIF1an-mediated AMPKα1 hydroxylation, miR-455-3p, activated AMPKα1 and triggered PGC1α phosphorylation leading to stimulation of mitochondrial biogenesis (Figure 4). Another hypothesis behind the protective role of miR-455-3p in mitochondria by targeting mitogen-activated protein kinase kinase 1 (MEKK1) gene (Wang and Shen, 2022), which is well known for the transition of mitochondrial permeability leading to apoptosis (Gibson et al., 2002; Mendoza et al., 2005). A binding correlation between miR-455-3p and MEKK1 was initially found in bioinformatics analysis and then validated by dual luciferase and RIP (RNA-Protein pull-down) assays. miR-455-3p specifically targeted and lowered MEKK1 expression also in in-vitro cell culture (Wang and Shen, 2022).
Figure 4:

Role of miR455-3p in mitochondrial biogenesis and synaptic architecture. miRNA 455-3p repress HIF1an expression and improves mitochondrial biogenesis. Our hypothesis is miRNA improved dendritic arbor and synaptic strength by positively regulating mitochondrial function and oxidative stress.
e. miR-455-3p enhanced dendritic and synaptic architecture
miR-455-3p has a significant role in promoting neuronal growth, supporting dendritic architecture, developing spines and augmenting synaptogenesis. At first, we observed a significant increase in synaptic proteins like synaptophysin and PSD95 in neuroblastoma cells (Kumar et al., 2019a) supporting its role in the regulation of synaptic activities. Moreover, miR-455-3p showed protective effects against AD related synaptic damages as synaptic protein levels were increased significantly in miR-455-3p co-transfected cells compared to the cells transfected with mutant APP alone. In our miR-455-3p TG mouse model, miR-455-3p overexpression increased neuronal populations, dendritic lengths and density, number of spines in hippocampal segments of TG mice brain (Kumar et al., 2021a). Furthermore, average synapse numbers were significantly upregulated in TG mice compared to WT. Findings from our TG and KO mice models are elaborated in the letter section of this article.
Several other articles reported the role of miR-455-3p in protecting neuronal survival against neurotoxicity. First, Yao et al., reported that miR-455 had a vital role in protecting neuronal cell death by downregulating TNF Receptor Associated Factor 3 (TRAF3) in cerebral ischemic stroke (Yao et al., 2016). Second, high levels of miR-455-3p reduces the propofol-induced neurotoxicity by suppressing the EphA4 (ephrin Type-A Receptor 4) expression in hippocampal neurons (Zhu et al., 2020). Third, miR-455-3p ameliorated hippocampal neuronal injury by conversely regulating programmed cell death 7 (PDCD7) expression (Gan and Ouyang, 2022). Though these findings strongly suggest its protective role in progression and pathogenesis of AD, further research is still required to understand the underlying mechanism(s).
f. miR-455-3p mice models showed superior cognitive ability and extended longevity
Elevated amount of miR-455-3p have been shown to increase synaptic activity, mitochondrial function and biogenesis, and decrease Aβ pathology, based on both in vitro and in vivo studies in our lab (Kumar et al., 2021b). The widely accepted models for examining the physiological processes and functions of miRNAs in the progression of AD are mice. Consequently, we present a thorough summary of the implications of miR-455-3p on cognitive behaviour, mitochondrial activity, and synaptic activity by investigating our miR-455-3p KO and TG mice models.
A number of AD mice models, including KO, knock-in, and TG models, have been employed to both produce therapeutic medications and comprehend the disease process from the point of birth to the end. These models allow researchers to look into the effects of gene products and their genetic mutations, and to evaluate anti-AD medications by crossing them with other TG and KO mice (Elder et al., 2010; Hall and Roberson, 2012). A innovative method for examining the advantageous or detrimental impacts of miRNAs in human diseases is the use of miRNA mouse models (Delay and Hébert, 2011; Pal and Kasinski, 2017). A single miRNA, such as miR-455-3p, can control several target proteins, which is why it is linked to a number of human diseases, such as chondrogenesis and cancer (Kumar and Reddy, 2019). Because of miR-455-3p’s important functions in AD and possible anti-AD benefits, the development of miR-455-3p mice models has become imperative. In addition to AD, miR-455-3p mice models could improve knowledge about and therapy for additional human disorders linked to miR-455-3p.
It has been discovered that miRNA molecules profoundly affected endogenous human genes and proteins on an epigenetic and genetic level (Bartel, 2004). Numerous miRNAs have been investigated in relation to AD, and their potential impacts on AD-related genes and proteins have been shown. In-depth investigation was hindered in the majority of miRNA studies due to the absence of appropriate mice models (Kim et al., 2015; Kumar et al., 2021b; Vilardo et al., 2010; Zhao et al., 2019). Numerous investigations used direct oral/nasal delivery of miRNA complexes or injections of miRNAs (Kumar et al., 2021b; Merhautova et al., 2016). The efficacy of these methods was restricted by various parameters, including the exogenous miRNA complex’s half-life within the body, immunological responses to miRNA-encapsulated particles, the stability of the miRNA complexes, and the route and dosage of miRNA administration. The creation of miRNA mice models, which offer comprehensive insights into the physiological mechanisms and therapeutic potential of particular miRNAs, is perfect for overcoming these obstacles and technological constraints (Kumar et al., 2019b; Kumar and Reddy, 2018b, 2019, 2021; Kumar et al., 2017b). Based on earlier studies into miR-455-3p and its prospective anti-AD capabilities, miR-455-3p KO and TG models were generated effectively by employing CRISPR/Cas9 knockout procedures and pronuclear injection of the miR-455-3p transgene into mice embryos (Figure 5). miR-455-3p expression levels were examined by immunoblotting, qRT-PCR, sequencing, and genotyping in TG and KO mice. In these animal models, the insertion or deletion of the miR-455-3p transgene did not result in notable phenotypic abnormalities (Kumar et al., 2021b).
Figure 5:

Unveiling the role of different mice models of miR-455-3p in diverse physiological processes.
Our study tracks non-KO and non-transgenic WT, TG, and KO mice from birth to the end of their lives, which provides an excellent explanation for the extinction of life span. According to survival data, age-matched transgenic mice outlived their non-transgenic WT siblings by 5 months, but KO mice outlived their WT siblings by 4 months (Kumar et al., 2021b). This explains why miR-455-3p overexpression increases longevity; put another way, miR-455-3p has brain-defending attributes. Given that naturally generated whole APP protein and mRNA levels were higher in KO mice and lower in miR-455-3p TG animals, the findings imply that reducing full-length APP may have a protective effect on the brain (Kumar et al., 2021b; Swingler et al., 2022). The significant rise in endogenous APP levels in KO mice’s brains suggests the value of using KO animals to investigate APP-related toxicity in AD. Crossing miR-455-3p TG/KO mice with other AD mice model is essential to ascertain whether miR-455-3p can lessen the toxicities of full-length APP in mice (Kumar et al., 2021b). This is especially true for mice with an unmodified 3’ UTR, such as the recently created humanized Ab-KI mice. The subsequently created double mutant animals, hAb-KI X TG miR-455-3p, were projected to be less dangerous to mitochondria and synaptic activity from APP and its c-terminal derivative, as well as to live longer and exhibit less cognitive decline. In contrast, it was projected that mice with double mutant hAb-KI X KO miR-455-3p would live shorter lives, experience more cognitive decline, and develop AD pathologies more quickly (Kumar et al., 2021b; Kumar et al., 2019b).
The main physiological indicator of AD etiology is cognitive dysfunction. Age-matched WT and TG mice fared better on the MWM (Morris Water Maze) test in terms of cognitive learning and memory, but KO mice did poorly. These findings imply that increased amount of endogenous miR-455-3p enhances cognitive abilities (Honer et al., 2019; John and Reddy, 2021; Kennedy, 2016; Kumar and Reddy, 2020). The brains and hippocampal regions of TG mice had noticeably more synapses in addition to greater amounts of the proteins MAP2, PSD95, and SNAP25. Furthermore, the hippocampal neurons of miR-455-3p TG mice exhibited higher dendritic spine density. It is yet unknown how miR-455-3p overexpression upregulates synaptic activity and proteins. It is commonly known that improving synaptic proteins and upregulating PGC1α improves cognitive function (Cheng et al., 2012; Honer et al., 2019). According to recent researches, cognitive performance can be enhanced by upregulating miR-455-3p by inhibiting EphA4 and HDAC2. Increased miR-455-3p levels lessen neurotoxicity by preventing hippocampus neurons from producing EphA4. Subsequent research highlighted the role of miR-455-3p in correcting neurological abnormalities and fixing memory impairments in TBI (traumatic brain injury) patients. It has been discovered that cinnamic acid reduces memory deficits in TBI patients by downregulating HDAC2 and upregulating the miR-455-3p expression levels (Guo et al., 2019; Zhu et al., 2020).
We also found that miR-455-3p directly improved neuronal differentiation. In TG mice, NeuN labelling demonstrated a notable rise in the neuronal population. Increased amount of the neural marker NeuN and dendritic marker MAP2 further supported the enlarged neuronal population caused by miR-455-3p overexpression. It is intriguing to note that the astrocyte population was subsequently lower in TG mice than in KO and WT animals. As expected, KO mice with miR-455-3p knockdown showed decreased neuronal population and increased expressions of astrocytes and microglias (Kumar et al., 2021b). The development of abnormal mitochondrial function and structure is a major source of synaptic dysfunction in AD. miR-455-3p affected the shape and function of mitochondria, according to previous studies (Calkins and Reddy, 2011; Reddy, 2011). The increase in mitochondrial length and number in TG mice demonstrated improved mitochondrial organization, indicating higher mitochondrial quality, according to our TEM analysis. An increase in the quantity of proteins involved in mitochondrial biogenesis enforced the improvement. The TG mice showed positively regulated dynamics proteins (MFN2, MFN1, FIS1, DRP1, and OPA1) and elevated expressions of mitochondrial biogenesis proteins (NRF2, TFAM, NRF1, and PGC1α). However, there are several opinions on how miR-455-3p affects mitochondrial activities. miR-455-3p’s regulation of HIF1α is one of the recognized explanations (Zhang et al., 2015). Moreover, the correlation between HIF1α and PGC1α in controlling mitochondrial activity indicates that miR-455-3p functions as a key mediator of mitochondrial activity in TG mice by way of HIF1α and PGC1α (O’Hagan et al., 2009; Yun et al., 2019).
In conclusion, miR-455-3p improves brain health generally and has a good effect on neuronal activity via a variety of mechanisms. More genetic crossover research employing APP TG animal lines and hAb-KI is required to completely comprehend miR-455-3p’s protective actions against mutant APP proteins. These animals offer significant opportunities for studying the physiological pathways of miR-455-3p in AD and other human disorders.
1.3. miR-455-3p as a potential peripheral biomarker for AD
Based on the above findings and discussion, miR455-3-p emerges as a potential candidate to be considered as biomarker for AD. Here, we discuss on the role of different miRNAs as biomarkers for AD, Reddy lab findings for miR455-3-p as a biomarker, and a scheme for developing miR-455-3p Kit for AD and MCI diagnosis.
a. MicroRNAs role as a biomarker in AD
It has been demonstrated that microRNAs, which are tiny non-coding RNAs, control the expression of certain genes. Changes in miRNA expression have been linked to various pathological processes, including neurodegeneration (Femminella et al., 2015). In the search for easily accessible, non-invasive biomarkers for AD diagnosis and prognosis, circulating miRNAs are among the most promising candidates (Femminella et al., 2015). Several of them have been repeatedly found in AD, and their targets likewise appear to be involved in the pathophysiological mechanisms that underlie AD. Certain miRNAs were expressed in the central nervous system (CNS), where they control neurite outgrowth, synaptic plasticity, and neuronal development. Except for the hippocampus, Landgraf et al. (2007) showed that human adult brain regions could be distinguished from adult rat brain regions using cluster analysis based on the expression of orthologous miRNAs. Additionally, adult and embryonic brain tissues form distinct clusters (Landgraf et al., 2007). Several miRNAs potentially related AD pathology have been consistently identified as dysregulated in AD (Van den Hove et al., 2014). For example, miR-455, miR-191, miR-106, miR-146a, miR-29, miR-34, miR-107, miR-181 etc. One of the most commonly changed miRNAs in AD is miR-9, which is encoded by three distinct genes. It has been demonstrated that adding Aβ peptides to primary neuronal cell cultures would downregulate this tiny RNA, which is abundantly expressed in the prenatal hippocampal region and downregulated in AD brains (Krichevsky et al., 2003). Early in AD, there has been evidence that the short RNA miR-107 is downregulated in the temporal cortex. Its expression has been shown to target BACE1, hence controlling the synthesis of amyloid, and it also has a negative connection with neuritic plaque density and neurofibrillary tangles (Nelson and Wang, 2010).
The miR-29 family of miRNAs has been demonstrated to enhance amyloid formation in vitro neuronal models and exhibit an inverse correlation with BACE1 (Hébert et al., 2008). The study showed downregulation in the cerebellum, temporal cortex, and patient serum of human AD patients (Hébert et al., 2008). On the other hand, miR-34 controls p53 expression in AD, which is linked to tau phosphorylation (Hooper et al., 2007). Moreover, transgenic mice models of AD and the hippocampi of AD patients both exhibit elevated levels of miR-34 (Zovoilis et al., 2011). Whereas miR-181 functions as a gene regulator for oncogene RAS and tumor necrosis factor-alpha. Aβ has the ability to downregulate miR-181 expression in primary human astrocyte cultures, and in vitro tests have demonstrated a correlation between miR-181 and Aβ levels in the human AD brain (Schonrock et al., 2012). It has been demonstrated that miR-106a and miR-106b directly bind to APP mRNA and are downregulated in AD patients’ temporal cortical regions (Kim et al., 2012). In addition to the miRNAs mentioned earlier, others including miR-124, miR-132, miR-153, and miR-146a have also been linked to AD and other neurodegenerative conditions. Some of them appear to be engaged in more than one of these processes, indicating that they may also mediate cross-talk among the various pathogenic processes underlying AD. The majority of them have demonstrated significance in tau phosphorylation, neuroinflammation, APP processing, and ApoE lipidization (Sethi and Lukiw, 2009) (Goodall et al., 2013; Van den Hove et al., 2014).
Mild cognitive impairment (MCI) is characterized as a diverse clinical syndrome marked by notable changes in cognitive function and deficits identified through neuropsychological testing, while overall daily functionality remains relatively intact (Winblad et al., 2004). The diagnostic criteria for MCI have evolved over time (Ogonowski et al., 2022). Finding novel biomarkers for the diagnosis and prognosis of MCI is a major task. Within this context, serum, plasma, and other bodily fluids’ miRNAs have become a potentially useful source of biomarkers for cognitive deficits associated with MCI and AD. It is interesting to note that, in response to pathophysiological stimuli, miRNAs can control various signaling pathways via a variety of targets (Ogonowski et al., 2022). The “miR-134 family” (miR-134/miR-370, miR-323-3p/miR-370, and miR-382/miR-370) as well as the “miR-132 family” (miR-128/miR-491-5p, miR-132/miR-491-5p, and mir-874/miR-491-5p) comprise the identified biomarker pairs (Sheinerman et al., 2013). miRNAs such as miR-27b-3p, miR-92a-3p, miR-181c-5p, miR-181a-5p, miR-210-3p, and miR-455-3p that have been connected to AD and MCI, have been demonstrated to be elevated, whereas miR-93-5p, miR-107, miR-127-3p, miR-142-3p, and miR-363-3p, miR-15b-3p, miR-22-5p, miR-29b-3p, miR-30c-5p, and miR-30b-5p were among the 67% of miRNAs that were downregulated. This finding raises the possibility that these 16 miRNAs are predictive biomarkers for MCI (Ogonowski et al., 2022). Considering these, the role of miRNAs as biomarkers in AD and MCI has been a rapidly growing area of research. Their ability to reflect underlying molecular changes, their presence in accessible biofluids, and their dynamic expression profiles make them valuable tools for early diagnosis, prognosis, and therapeutic monitoring. As research advances, the integration of miRNA biomarkers into clinical practice holds the promise of improved management and treatment of AD and MCI, ultimately enhancing patient outcomes.
b. miR-455-3p as a biomarker for AD
The Reddy lab has discovered miR-455-3p as a viable peripheral biomarker for AD (Kumar and Reddy, 2019). We (Reddy lab) measured miRNAs in serum samples from persons with MCI and AD in comparison to healthy controls in order to accomplish our goal. Affymetrix microarray analysis was utilized, and qRT-PCR was employed to confirm the differentially expressed miRNAs. Furthermore, utilizing AD cell lines, AD fibroblasts, AD cerebrospinal fluids, transgenic mice for the amyloid precursor protein, AD B-lymphocytes (all from late-onset AD; LOAD), and postmortem brains from AD patients, we found that AD cases had higher expression levels of miR-455-3p than samples from healthy controls. We further confirmed the miRNA results. We observed a progressive increase in the expression of four miRNAs: miR-455-3p, miR-4668-5p, miR-3613-3p, and miR-4674. When compared to controls, those with AD and mild cognitive impairment had lower levels of mir-6722. Among them, qRT-PCR validation analysis revealed that only miR-455-3p (P = 0.007) and miR-4668-5p (P = 0.016) were significantly upregulated in AD patients as compared to healthy controls. When compared to controls, it revealed an astonishing 11.3-fold increase in expression for miR-455-3p in AD patients. Additionally, miR-455-3p was shown to be upregulated (P = 0.016) in the AD postmortem brains with varying Braak stages according to qRT-PCR analysis. Nevertheless, A significant area under the curve (AUC) value was only found for miR-455-3p in the serum (AUROC = 0.79; P = 0.015) and brains (AUROC = 0.86; P = 0.016) of AD patients, according to receiver operating characteristic curves (ROC) curve analysis. Again, the cerebral cortex, the region of the brain affected by AD, had a 1.8-fold high level of miR-455-3p (P = 0.004), while the cerebellum, the non-affected area, had a low level of this protein, according to an expression study of transgenic mice for amyloid precursor protein. Moreover, miR-455-3p was equally overexpressed in the Aβ (1–42) peptide treatment of neuroblastoma cells in humans and mice. The Aβ-treated SH-SY5Y cells exhibited a 4.1-fold (P = 0.027) increase in hsa-miR-455-3p expression in comparison to the control (untreated) cells (Figure 6). Likewise, in N2a cells, the expression of miR-455-3p was elevated by 3.8-fold (P = 0.021) in Aβ-treated cells as opposed to control cells. Functional investigation using the miR-path on differentially expressed miRNAs revealed that miR-455-3p was involved in the control of multiple biological pathways.
Figure 6:

This image illustrates the elevated expression levels of miR-455-3p in AD related specimes; serum and CSF collected from AD and MCI patients, postmortem AD brain sections, and AD cell culture models.
It was discovered that genes connected to these pathways play a critical role in the pathophysiology of AD. Several signaling networks and a number of AD-related genes have been directly connected to the pathophysiology of the disease. The link between miR-455-3p and 11 biological pathways and related genes was determined by the miRNA analysis. The TGF-β signaling pathway, the hippo signaling pathway, the ECM-receptor interaction, and the control of the actin cytoskeleton were the most significant signaling pathways (Kumar et al., 2017b). Another study from Reddy’s lab (Kumar and Reddy, 2018b) reported that, compared to healthy control subjects, fibroblasts and B-lymphocytes from AD patients had higher levels of miR-455-3p. Notably, sporadic AD patients’ cells had much higher levels of miR-455-3p than did the healthy control’s cells. In particular, B-lymphocytes from sporadic AD individuals had considerably greater levels of miR-455-3p controls (P = 0.044). An examination of the Receiver Operating Characteristic (ROC) curve showed that AD fibroblast cells (AUROC = 0.861; P = 0.03) and postmortem brains (AUROC = 0.792; P = 0.001) had a substantial area under the curve (AUROC) value for miR-455-3p. These results clearly suggest that miR-455-3p is a potential biomarker for AD (Kumar and Reddy, 2018b). However, more research is required to elucidate the molecular connections between miR-455-3p and the progression of AD. From these findings, we can infer that miR-455-3p might serve as a potential peripheral biomarker for diagnosing AD and MCI. While numerous studies have suggested miRNAs as potential markers for AD, no one miRNA has yet to be clinically verified as a reliable biomarker. The reported findings assist in identifying a promising candidate, with the expression levels of miR-455-3p suggesting its potential role as a peripheral biomarker (Kumar et al., 2017b).
c. Development of a miR-455-3p kit for MCI and AD diagnosis
Based on the above findings and discussions (Kumar and Reddy, 2018b; Kumar et al., 2017b), Dr. Reddy’s lab is developing a biochemical kit utilizing miR-455-3p for the diagnosis of AD and MCI. By developing this kit, we can detect MCI and AD in patients early, allowing for timely therapeutic interventions that can potentially slow the progression of these conditions, thereby improving patient outcomes and quality of life. Additionally, miR-455-3p can be measured in blood samples, making the diagnostic test minimally invasive compared to procedures like cerebrospinal fluid analysis or brain imaging. A blood-based test is more comfortable and less risky for patients, particularly the elderly, who may find other diagnostic procedures invasive and stressful. Focusing on a specific microRNA consistently altered in AD and MCI, the kit can provide a more sensitive and reliable diagnostic tool, reducing the chances of false positives and negatives. Early and accurate diagnosis of MCI and AD can also reduce healthcare costs by preventing or delaying the need for more intensive care and treatment as the disease progresses. Moreover, with a reliable diagnostic tool, healthcare resources can be better allocated to those who need them most, improving overall healthcare efficiency and patient care. In summary, the development of a miR-455-3p diagnostic kit is crucial for improving the early detection, diagnosis, and management of MCI and AD. It holds promise for enhancing patient outcomes and optimizing healthcare resources.
However, the results obtained from the miR-455-3p diagnostic kit should be corroborated with additional assessments to confirm the disease, including brain scans and the Montreal Cognitive Assessment (MoCA). The official scoring range for the MoCA is as follows: normal cognition - 26–30 points, mild cognitive impairment - 18–25 points, moderate cognitive impairment - 10–17 points, and severe cognitive impairment - under 10 points. Additionally, it is important to test for cell-free DNA and DNA double-strand breakage to get a comprehensive diagnostic profile.
For the development of the miR-455-3p diagnostic kit, Dr. Reddy’s lab proposes the following steps:
Blood Collection: Collect blood samples from patients.
Serum Preparation: Separate the serum from the blood and store it at the appropriate temperature.
RNA Isolation and cDNA Synthesis: Isolate total RNA from the serum, perform polyadenylation, and synthesize cDNA using miR-455-3p specific primers.
Gene Expression Analysis: Normalize and quantify gene expression. Calculate miR-455-3p fold changes using quantitative reverse transcription–polymerase chain reaction (RTqPCR). (Figure: 6).
Additionally, the miR-455-3p levels will be validated using fluorescence spectra, following the procedures described by Guo and colleagues (Guo et al., 2022).
It is anticipated that the fluorescence spectral analysis of miR-455-3p will authenticate the miR-455-3p data and increase the accuracy of AD diagnosis. In this process, a dual-signal DNA probe based on the fluorescence recovery of carbon quantum dots (CQDs) and the sulfo-cyanine5 (Cy5) dye will be used to determine miR-455-3p simultaneously. In order to amplify the signals through a cycle of DNA degradation and RNA re-hybridization, duplex-specific nuclease will be added. The method’s dynamic ranges were 0.01 to 4 pM for miR-455-3p and 0.01 to 5 pM for that same compound. The recovery of miRNAs in spiking human serum and its strong correlation with RT-qPCR testing will validate the method’s accuracy. All enrolled individuals, including healthy donors, MCI patients, and AD patients, will have their blood samples tested to measure the miRNA content. However, a straightforward and sensitive technique for the very accurate simultaneous measurement of miRNAs has been published, and it suggests that the concentration ratio of miR-455-3p is a useful blood indicator for the early and in-stage diagnosis of AD (Guo et al., 2022).
Conclusions and Future Directions
An ideal biomarker for diagnosing MCI and AD progression should be early detectable, non-invasive in nature, preferably peripherally available. We reported a significant upregulation of mir-455-3-p expression in AD postmortem brains, AD fibroblasts, and B-lymphocytes. We also found high level of mir-455-3-p in AD serum, which was later validated by the presence of an increased miR-455-3p in CSF exosomes secreted from the brain. miR-455-3p targets many genes related to oxidative decay, mitochondrial dysfunction, synaptic damage and neurodegeneration process in AD pathogenesis. More importantly, miR-455-3p dramatically decreases full-length APP expression, the C-terminal fragment of APP and reduces Aβ(1–40) and Aβ(1–42) levels. Our in-vitro cell culture studies strongly explains the role of miR-455-3p in protecting neuronal cells against AD related stress including mutant APP. Moreover, mir-455-3-p mice models showed its vibrant role in maintaining and promoting memory and cognitive functions in an extended life period. Overall, its positive role in the regulation of mitochondrial biogenesis/dynamics and synaptic activity make it worthy of research to identify and validate AD diagnosis and our proposed miR-455-3p detection kit will serve the purpose largely.
Figure 7:

Diagram showing the progression of the miR-455-3p detection kit development: Twenty milliliters of blood will be drawn, and it will be processed right away to separate the buffy coat and serum components. Following the isolation of total RNA, a set of procedures will be carried out, including polyadenylation and the synthesis of cDNA using primers specific to miR-455-3p. The study of fluorescence spectra will be used to confirm the correctness of miR-455-3p levels.
Highlights.
MicroRNAs are small non-coding RNAs evolutionary conserved molecules that are involved with Alzheimer’s disease.
MicroRNAs regulate cellular processes, including RNA silencing, post-translational gene expression and neurodegeneration.
Overexpression of miR-455-3p in mice showed superior cognitive learning, improved memory and extended lifespan.
Reddy lab research revealed that microRNA-455-3P is a potential biomarker and therapeutic target for AD.
Funding:
The research presented in this article was supported by NIH grant AG079264 to PHR.
Abbreviations:
- ACVR2B
Activin Receptor Type IIB
- AD
Alzheimer’s disease
- ADRD
Alzheimer’s disease related disease
- AGO
Argonaute
- AMPK α1
AMP-activated kinase α1 subunit
- APHA1
Aph-1 Homolog A
- APOE4
apolipoprotein E4
- APP
Amyloid precursor protein
- ARE
Antioxidant response elements
- Aβ
beta-amyloid
- BACE1
Beta-site APP cleaving enzyme-1
- BMP4
Bone morphogenetic protein 4
- CD2AP
CD2 Associated Protein
- CHRDL1
Chordin-like 1
- CNS
Central nervous system
- COL27A1
collagen type XXVII alpha 1 chain
- CQDs
Carbon quantum dots
- CSF
Cerebrospinal fluid
- Cy5
Cyanine5
- DGCR8
DiGeorge Syndrome Critical Region 8
- DLD
Dihydrolipoamide dehydrogenase
- DNA
Deoxyribonucleic acid
- EPHA4
Ephrin Type-A Receptor 4
- HDAC2
Histone deacetylases 2
- HIF1an
hypoxia-inducible factor 1 α inhibitor
- Hmgb1
High-mobility group box
- IPF
Idiopathic pulmonary fibrosis
- KO
Knockout
- MCI
Mild cognitive impairment
- MEKK1
Mitogen-activated protein kinase kinase 1
- mir-455-3-p
MicroRNA-455-3-p
- miRISC
miRNA-induced silencing complex
- MoCA
Montreal cognitive assessment
- mRNA
Messenger RNA
- MS4A4
membrane-spanning 4-domains subfamily A
- ncRNA
Noncoding RNAs
- NFTs
Neurofibrillary tangles
- NGF
Nerve growth factor
- NRF2
nuclear factor erythroid 2-related factor 2
- OGD
Oxygen and glucose deprivation
- PAK2
P21-activated kinase 2
- PDCD7
Programmed cell death 7
- pre-miRNAs
precursor-miRNAs
- pri-miRNAs
primary miRNAs
- PS1
presenilin 1
- PS2
presenilin 2
- PTEN
Phosphatase and tensin homolog
- RISC
RNA-induced silencing complex
- RNA
Ribonucleic acids
- ROC
Receiver operating characteristic
- RT-qPCR
Quantitative reverse transcription–polymerase chain reaction
- SNHG15
Small nucleolar RNA host gene 15
- SOX2-OT
Sex-determining region Y-box 2 overlapping transcript
- TBI
Traumatic brain injury
- TG
Transgenic
- TGF-β
Transforming growth factor beta
- TP53INP1
Tumor protein p53 inducible nuclear protein 1
- TRAF3
TNF Receptor Associated Factor 3
- TRAF6
Tumor necrosis factor receptor associated factor 6
- TβRI
TGF-β type I receptor
- TβRII
TGF-β type II receptors
- UTR
Untranslated region
- ZEB1
Zinc finger E-box-binding homeobox 1
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Conflicts of Interest: The authors declare no conflict of interest.
References:
- Amaral PP, Dinger ME, Mercer TR, Mattick JS, 2008. The eukaryotic genome as an RNA machine. science 319, 1787–1789. [DOI] [PubMed] [Google Scholar]
- Ambros V, 2004. The functions of animal microRNAs. Nature 431, 350–355. [DOI] [PubMed] [Google Scholar]
- Arai T, Kojima S, Yamada Y, Sugawara S, Kato M, Yamazaki K, Naya Y, Ichikawa T, Seki N, 2019. Pirin: a potential novel therapeutic target for castration-resistant prostate cancer regulated by miR-455-5p. Molecular oncology 13, 322–337. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bartel DP, 2004. MicroRNAs: genomics, biogenesis, mechanism, and function. cell 116, 281–297. [DOI] [PubMed] [Google Scholar]
- Calkins MJ, Reddy PH, 2011. Amyloid beta impairs mitochondrial anterograde transport and degenerates synapses in Alzheimer’s disease neurons. Biochimica et Biophysica Acta (BBA)-Molecular Basis of Disease 1812, 507–513. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Caraci F, Battaglia G, Busceti C, Biagioni F, Mastroiacovo F, Bosco P, Drago F, Nicoletti F, Sortino MA, Copani A, 2008. TGF-β1 protects against Aβ-neurotoxicity via the phosphatidylinositol-3-kinase pathway. Neurobiology of disease 30, 234–242. [DOI] [PubMed] [Google Scholar]
- Carthew RW, Sontheimer EJ, 2009. Origins and mechanisms of miRNAs and siRNAs. Cell 136, 642–655. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cech TR, Steitz JA, 2014. The noncoding RNA revolution—trashing old rules to forge new ones. Cell 157, 77–94. [DOI] [PubMed] [Google Scholar]
- Chalmers KA, Love S, 2007. Neurofibrillary tangles may interfere with Smad 2/3 signaling in neurons. Journal of Neuropathology & Experimental Neurology 66, 158–167. [DOI] [PubMed] [Google Scholar]
- Chen Y-G, 2009. Endocytic regulation of TGF-β signaling. Cell research 19, 58–70. [DOI] [PubMed] [Google Scholar]
- Cheng A, Wan R, Yang J-L, Kamimura N, Son TG, Ouyang X, Luo Y, Okun E, Mattson MP, 2012. Involvement of PGC-1α in the formation and maintenance of neuronal dendritic spines. Nature communications 3, 1250. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cheng CM, Shiah SG, Huang CC, Hsiao JR, Chang JY, 2016. Up-regulation of miR-455-5p by the TGF-β–SMAD signalling axis promotes the proliferation of oral squamous cancer cells by targeting UBE2B. The Journal of pathology 240, 38–49. [DOI] [PubMed] [Google Scholar]
- Chopra N, Wang R, Maloney B, Nho K, Beck JS, Pourshafie N, Niculescu A, Saykin AJ, Rinaldi C, Counts SE, 2021. MicroRNA-298 reduces levels of human amyloid-β precursor protein (APP), β-site APP-converting enzyme 1 (BACE1) and specific tau protein moieties. Molecular psychiatry 26, 5636–5657. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Christen Y, 2000. Oxidative stress and Alzheimer disease. The American journal of clinical nutrition 71, 621S–629S. [DOI] [PubMed] [Google Scholar]
- DeKosky ST, Scheff SW, Styren SD, 1996. Structural correlates of cognition in dementia: quantification and assessment of synapse change. Neurodegeneration 5, 417–421. [DOI] [PubMed] [Google Scholar]
- Delay C, Hébert SS, 2011. MicroRNAs and Alzheimer’s disease mouse models: current insights and future research avenues. International journal of Alzheimer’s disease 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dhandapani KM, Hadman M, De Sevilla L, Wade MF, Mahesh VB, Brann DW, 2003. Astrocyte protection of neurons: role of transforming growth factor-beta signaling via a c-Jun-AP-1 protective pathway. J Biol Chem 278, 43329–43339. [DOI] [PubMed] [Google Scholar]
- Du H, Guo L, Yan S, Sosunov AA, McKhann GM, ShiDu Yan S, 2010. Early deficits in synaptic mitochondria in an Alzheimer’s disease mouse model. Proceedings of the National Academy of Sciences 107, 18670–18675. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Elder GA, Gama Sosa MA, De Gasperi R, 2010. Transgenic mouse models of Alzheimer’s disease. Mount Sinai Journal of Medicine: A Journal of Translational and Personalized Medicine: A Journal of Translational and Personalized Medicine 77, 69–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Eulalio A, Huntzinger E, Izaurralde E, 2008. Getting to the root of miRNA-mediated gene silencing. Cell 132, 9–14. [DOI] [PubMed] [Google Scholar]
- Fan Y, Wei L, Zhang S, Song X, Yang J, He X, Zheng X, 2021. LncRNA SNHG15 knockdown protects against OGD/R-induced neuron injury by downregulating TP53INP1 expression via binding to miR-455-3p. Neurochemical research 46, 1019–1030. [DOI] [PubMed] [Google Scholar]
- Femminella GD, Ferrara N, Rengo G, 2015. The emerging role of microRNAs in Alzheimer’s disease. Frontiers in physiology 6, 130339. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gan C, Ouyang F, 2022. Exosomes Released from Bone-Marrow Stem Cells Ameliorate Hippocampal Neuronal Injury Through transferring miR-455-3p. Journal of Stroke and Cerebrovascular Diseases 31, 106142. [DOI] [PubMed] [Google Scholar]
- Gao X, Zhao H, Diao C, Wang X, Xie Y, Liu Y, Han J, Zhang M, 2018. miR-455-3p serves as prognostic factor and regulates the proliferation and migration of non-small cell lung cancer through targeting HOXB5. Biochemical and biophysical research communications 495, 1074–1080. [DOI] [PubMed] [Google Scholar]
- Gharanei S, Shabir K, Brown JE, Weickert MO, Barber TM, Kyrou I, Randeva HS, 2020. Regulatory microRNAs in brown, brite and white adipose tissue. Cells 9, 2489. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gibson EM, Henson ES, Villanueva J, Gibson SB, 2002. MEK kinase 1 induces mitochondrial permeability transition leading to apoptosis independent of cytochrome c release. J Biol Chem 277, 10573–10580. [DOI] [PubMed] [Google Scholar]
- Goodall EF, Heath PR, Bandmann O, Kirby J, Shaw PJ, 2013. Neuronal dark matter: the emerging role of microRNAs in neurodegeneration. Frontiers in cellular neuroscience 7, 178. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gu Q, Wang B, Zhao H, Wang W, Wang P, Deng Y, 2020. LncRNA promoted inflammatory response in ischemic heart failure through regulation of miR-455-3p/TRAF6 axis. Inflammation Research 69, 667–681. [DOI] [PubMed] [Google Scholar]
- Guo S, Zhen Y, Zhu Z, Zhou G, Zheng X, 2019. Cinnamic acid rescues behavioral deficits in a mouse model of traumatic brain injury by targeting miR-455-3p/HDAC2. Life sciences 235, 116819. [DOI] [PubMed] [Google Scholar]
- Guo Y, Liu D, Yang Q, Shi W, Yang Z, Chen J, Xiang J, 2022. Early and in-stage diagnosis of Alzheimer’s disease by the simultaneous fluorescent determination of the biomarkers miR-501-3p and miR-455-3p with carbon quantum dots in serum. Analytical Letters 55, 1453–1465. [Google Scholar]
- Hall AM, Roberson ED, 2012. Mouse models of Alzheimer’s disease. Brain research bulletin 88, 3–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hébert SS, Horré K, Nicolaï L, Papadopoulou AS, Mandemakers W, Silahtaroglu AN, Kauppinen S, Delacourte A, De Strooper B, 2008. Loss of microRNA cluster miR-29a/b-1 in sporadic Alzheimer’s disease correlates with increased BACE1/β-secretase expression. Proceedings of the National Academy of Sciences 105, 6415–6420. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Honer WG, Ramos-Miguel A, Alamri J, Sawada K, Barr AM, Schneider JA, Bennett DA, 2019. The synaptic pathology of cognitive life. Dialogues in Clinical Neuroscience 21, 271–279. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hooper C, Meimaridou E, Tavassoli M, Melino G, Lovestone S, Killick R, 2007. p53 is upregulated in Alzheimer’s disease and induces tau phosphorylation in HEK293a cells. Neuroscience letters 418, 34–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hu S, Zhao X, Mao G, Zhang Z, Wen X, Zhang C, Liao W, Zhang Z, 2019. MicroRNA-455-3p promotes TGF-β signaling and inhibits osteoarthritis development by directly targeting PAK2. Experimental & Molecular Medicine 51, 1–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Huang WJ, Zhang X, Chen WW, 2016. Role of oxidative stress in Alzheimer’s disease. Biomed Rep 4, 519–522. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Huntzinger E, Izaurralde E, 2011. Gene silencing by microRNAs: contributions of translational repression and mRNA decay. Nature Reviews Genetics 12, 99–110. [DOI] [PubMed] [Google Scholar]
- Iwakawa H. o., Tomari Y, 2015. The functions of microRNAs: mRNA decay and translational repression. Trends in cell biology 25, 651–665. [DOI] [PubMed] [Google Scholar]
- Jacob F, Monod J, 1961. Genetic regulatory mechanisms in the synthesis of proteins. Journal of molecular biology 3, 318–356. [DOI] [PubMed] [Google Scholar]
- John A, Reddy PH, 2021. Synaptic basis of Alzheimer’s disease: Focus on synaptic amyloid beta, P-tau and mitochondria. Ageing research reviews 65, 101208. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kennedy MB, 2016. Synaptic signaling in learning and memory. Cold Spring Harbor perspectives in biology 8, a016824. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kim J, Yoon H, Horie T, Burchett JM, Restivo JL, Rotllan N, Ramírez CM, Verghese PB, Ihara M, Hoe H-S, 2015. microRNA-33 regulates ApoE lipidation and amyloid-β metabolism in the brain. Journal of Neuroscience 35, 14717–14726. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kim J, Yoon H, Ramírez CM, Lee S-M, Hoe H-S, Fernández-Hernando C, Kim J, 2012. MiR-106b impairs cholesterol efflux and increases Aβ levels by repressing ABCA1 expression. Experimental neurology 235, 476–483. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kosaka N, Iguchi H, Ochiya T, 2010. Circulating microRNA in body fluid: a new potential biomarker for cancer diagnosis and prognosis. Cancer science 101, 2087–2092. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kosaka N, Yoshioka Y, Hagiwara K, Tominaga N, Ochiya T, 2013. Functional analysis of exosomal microRNA in cell–cell communication research. Circulating MicroRNAs: Methods and Protocols, 1–10. [DOI] [PubMed] [Google Scholar]
- Krichevsky AM, King KS, Donahue CP, Khrapko K, Kosik KS, 2003. A microRNA array reveals extensive regulation of microRNAs during brain development. Rna 9, 1274–1281. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Krol J, Loedige I, Filipowicz W, 2010. The widespread regulation of microRNA biogenesis, function and decay. Nature reviews genetics 11, 597–610. [DOI] [PubMed] [Google Scholar]
- Kumar S, Morton H, Sawant N, Orlov E, Bunquin LE, Pradeepkiran JA, Alvir R, Reddy PH, 2021a. MicroRNA-455-3p improves synaptic, cognitive functions and extends lifespan: Relevance to Alzheimer’s disease. Redox Biol 48, 102182. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kumar S, Morton H, Sawant N, Orlov E, Bunquin LE, Pradeepkiran JA, Alvir R, Reddy PH, 2021b. MicroRNA-455-3p improves synaptic, cognitive functions and extends lifespan: Relevance to Alzheimer’s disease. Redox biology 48, 102182. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kumar S, Reddy AP, Yin X, Reddy PH, 2019a. Novel MicroRNA-455-3p and its protective effects against abnormal APP processing and amyloid beta toxicity in Alzheimer’s disease. Biochimica et Biophysica Acta (BBA) - Molecular Basis of Disease 1865, 2428–2440. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kumar S, Reddy AP, Yin X, Reddy PH, 2019b. Novel MicroRNA-455-3p and its protective effects against abnormal APP processing and amyloid beta toxicity in Alzheimer’s disease. Biochimica et Biophysica Acta (BBA)-Molecular Basis of Disease 1865, 2428–2440. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kumar S, Reddy P, 2018a. MicroRNA-455-3p as a potential biomarker for Alzheimer’s disease: an update. Front Aging Neurosci 10: 41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kumar S, Reddy PH, 2016. Are circulating microRNAs peripheral biomarkers for Alzheimer’s disease? Biochimica et Biophysica Acta (BBA)-Molecular Basis of Disease 1862, 1617–1627. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kumar S, Reddy PH, 2018b. MicroRNA-455-3p as a potential biomarker for Alzheimer’s disease: an update. Frontiers in aging neuroscience 10, 41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kumar S, Reddy PH, 2019. A new discovery of MicroRNA-455-3p in Alzheimer’s disease. Journal of Alzheimer’s Disease 72, S117–S130. [DOI] [PubMed] [Google Scholar]
- Kumar S, Reddy PH, 2020. The role of synaptic microRNAs in Alzheimer’s disease. Biochimica et Biophysica Acta (BBA)-Molecular Basis of Disease 1866, 165937. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kumar S, Reddy PH, 2021. Elevated levels of MicroRNA-455-3p in the cerebrospinal fluid of Alzheimer’s patients: a potential biomarker for Alzheimer’s disease. Biochimica et biophysica acta. Molecular basis of disease 1867, 166052. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kumar S, Vijayan M, Bhatti J, Reddy P, 2017a. MicroRNAs as peripheral biomarkers in aging and age-related diseases. Progress in molecular biology and translational science 146, 47–94. [DOI] [PubMed] [Google Scholar]
- Kumar S, Vijayan M, Reddy PH, 2017b. MicroRNA-455-3p as a potential peripheral biomarker for Alzheimer’s disease. Human molecular genetics 26, 3808–3822. [DOI] [PMC free article] [PubMed] [Google Scholar]
- LaFerla FM, Green KN, Oddo S, 2007. Intracellular amyloid-β in Alzheimer’s disease. Nature Reviews Neuroscience 8, 499–509. [DOI] [PubMed] [Google Scholar]
- Landgraf P, Rusu M, Sheridan R, Sewer A, Iovino N, Aravin A, Pfeffer S, Rice A, Kamphorst AO, Landthaler M, 2007. A mammalian microRNA expression atlas based on small RNA library sequencing. Cell 129, 1401–1414. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lau P, Bossers K, Janky R.s., Salta E, Frigerio CS, Barbash S, Rothman R, Sierksma AS, Thathiah A, Greenberg D, 2013. Alteration of the micro RNA network during the progression of Alzheimer’s disease. EMBO molecular medicine 5, 1613–1634. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee H.g., Ueda M, Zhu X, Perry G, Smith MA, 2006. Ectopic expression of phospho-Smad2 in Alzheimer’s disease: uncoupling of the transforming growth factor-β pathway? Journal of neuroscience research 84, 1856–1861. [DOI] [PubMed] [Google Scholar]
- Lewis BP, Burge CB, Bartel DP, 2005. Conserved seed pairing, often flanked by adenosines, indicates that thousands of human genes are microRNA targets. cell 120, 15–20. [DOI] [PubMed] [Google Scholar]
- Li Z, Meng Q, Pan A, Wu X, Cui J, Wang Y, Li L, 2017a. MicroRNA-455-3p promotes invasion and migration in triple negative breast cancer by targeting tumor suppressor EI24. Oncotarget 8, 19455–19466. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li Z, Meng Q, Pan A, Wu X, Cui J, Wang Y, Li L, 2017b. MicroRNA-455-3p promotes invasion and migration in triple negative breast cancer by targeting tumor suppressor EI24. Oncotarget 8, 19455. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu Y, Lu F-A, Wang L, Wang Y-F, Wu C-F, 2021. Long non-coding RNA NEAT1 promotes pulmonary fibrosis by regulating the MicroRNA-455-3p/SMAD3 axis. Molecular Medicine Reports 23, 1–1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Long JM, Lahiri DK, 2011. MicroRNA-101 downregulates Alzheimer’s amyloid-β precursor protein levels in human cell cultures and is differentially expressed. Biochemical and biophysical research communications 404, 889–895. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Long JM, Maloney B, Rogers JT, Lahiri DK, 2019. Novel upregulation of amyloid-β precursor protein (APP) by microRNA-346 via targeting of APP mRNA 5′-untranslated region: Implications in Alzheimer’s disease. Molecular psychiatry 24, 345–363. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ludwig N, Leidinger P, Becker K, Backes C, Fehlmann T, Pallasch C, Rheinheimer S, Meder B, Stähler C, Meese E, 2016. Distribution of miRNA expression across human tissues. Nucleic acids research 44, 3865–3877. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ma X, Liu L, Meng J, 2017. RETRACTED: MicroRNA-125b promotes neurons cell apoptosis and Tau phosphorylation in Alzheimer’s disease. Elsevier. [DOI] [PubMed] [Google Scholar]
- Malone CD, Hannon GJ, 2009. Small RNAs as guardians of the genome. Cell 136, 656–668. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mao P, Reddy PH, 2011. Aging and amyloid beta-induced oxidative DNA damage and mitochondrial dysfunction in Alzheimer’s disease: implications for early intervention and therapeutics. Biochimica et Biophysica Acta (BBA)-Molecular Basis of Disease 1812, 1359–1370. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Massagué J, 1998. TGF-β signal transduction. Annual review of biochemistry 67, 753–791. [DOI] [PubMed] [Google Scholar]
- Massagué J, Seoane J, Wotton D, 2005. Smad transcription factors. Genes & development 19, 2783–2810. [DOI] [PubMed] [Google Scholar]
- Mattson MP, 2004. Pathways towards and away from Alzheimer’s disease. Nature 430, 631–639. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mendoza FJ, Henson ES, Gibson SB, 2005. MEKK1-induced apoptosis is mediated by Smac/Diablo release from the mitochondria. Biochemical and Biophysical Research Communications 331, 1089–1098. [DOI] [PubMed] [Google Scholar]
- Merhautova J, Demlova R, Slaby O, 2016. MicroRNA-based therapy in animal models of selected gastrointestinal cancers. Frontiers in Pharmacology 7, 216844. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Milosevic J, Pandit K, Magister M, Rabinovich E, Ellwanger DC, Yu G, Vuga LJ, Weksler B, Benos PV, Gibson KF, 2012. Profibrotic role of miR-154 in pulmonary fibrosis. American journal of respiratory cell and molecular biology 47, 879–887. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Morris KV, Mattick JS, 2014. The rise of regulatory RNA. Nature Reviews Genetics 15, 423–437. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Morton H, Kshirsagar S, Orlov E, Bunquin LE, Sawant N, Boleng L, George M, Basu T, Ramasubramanian B, Pradeepkiran JA, Kumar S, Vijayan M, Reddy AP,, Reddy PH, 2021. Defective mitophagy and synaptic degeneration in Alzheimer’s disease: Focus on aging, mitochondria and synapse. Free Radic Biol Med. 172,652–667. [DOI] [PubMed] [Google Scholar]
- Nelson PT, Wang W-X, 2010. MiR-107 is reduced in Alzheimer’s disease brain neocortex: validation study. Journal of Alzheimer’s Disease 21, 75–79. [DOI] [PMC free article] [PubMed] [Google Scholar]
- O’Hagan KA, Cocchiglia S, Zhdanov AV, Tambuwala MM, Cummins EP, Monfared M, Agbor TA, Garvey JF, Papkovsky DB, Taylor CT, 2009. PGC-1α is coupled to HIF-1α-dependent gene expression by increasing mitochondrial oxygen consumption in skeletal muscle cells. Proceedings of the National Academy of Sciences 106, 2188–2193. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ogonowski N, Salcidua S, Leon T, Chamorro-Veloso N, Valls C, Avalos C, Bisquertt A, Rentería ME, Orellana P, Duran-Aniotz C, 2022. Systematic review: microRNAs as potential biomarkers in mild cognitive impairment diagnosis. Frontiers in Aging Neuroscience 13, 807764. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Oliver DMA, Reddy PH, 2019. Molecular Basis of Alzheimer’s Disease: Focus on Mitochondria. J Alzheimers Dis. 72(s1):S95–S116. [DOI] [PubMed] [Google Scholar]
- Olivieri F, Capri M, Bonafè M, Morsiani C, Jung HJ, Spazzafumo L, Viña J, Suh Y, 2017. Circulating miRNAs and miRNA shuttles as biomarkers: Perspective trajectories of healthy and unhealthy aging. Mechanisms of Ageing and Development 165, 162–170. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ong J, Timens W, Rajendran V, Algra A, Spira A, Lenburg ME, Campbell JD, van den Berge M, Postma DS, van den Berg A, 2017. Identification of transforming growth factor-beta-regulated microRNAs and the microRNA-targetomes in primary lung fibroblasts. PloS one 12, e0183815. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pal AS, Kasinski AL, 2017. Animal models to study microRNA function. Advances in cancer research 135, 53–118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ponting CP, Oliver PL, Reik W, 2009. Evolution and functions of long noncoding RNAs. Cell 136, 629–641. [DOI] [PubMed] [Google Scholar]
- Prehn J, Bindokas VP, Jordán J, Galindo MF, Ghadge GD, Roos RP, Boise LH, Thompson CB, Krajewski S, Reed JC, 1996. Protective effect of transforming growth factor-beta 1 on beta-amyloid neurotoxicity in rat hippocampal neurons. Molecular pharmacology 49, 319–328. [PubMed] [Google Scholar]
- Qin L, Zhang Y, Lin J, Shentu Y, Xie X, 2016. MicroRNA-455 regulates migration and invasion of human hepatocellular carcinoma by targeting Runx2. Oncology reports 36, 3325–3332. [DOI] [PubMed] [Google Scholar]
- Rawat P, Sehar U, Bisht J, Selman A, Culberson J, Reddy PH,2022. Phosphorylated Tau in Alzheimer’s Disease and Other Tauopathies. Int J Mol Sci. 23(21),12841. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Reddy PH, 2006. Amyloid precursor protein-mediated free radicals and oxidative damage: Implications for the development and progression of Alzheimer’s disease. Journal of neurochemistry 96, 1–13. [DOI] [PubMed] [Google Scholar]
- Reddy PH, 2011. Abnormal tau, mitochondrial dysfunction, impaired axonal transport of mitochondria, and synaptic deprivation in Alzheimer’s disease. Brain research 1415, 136–148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Reddy PH, Beal MF, 2008. Amyloid beta, mitochondrial dysfunction and synaptic damage: implications for cognitive decline in aging and Alzheimer’s disease. Trends in molecular medicine 14, 45–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Reddy PH, Manczak M, Mao P, Calkins MJ, Reddy AP, Shirendeb U, 2010. Amyloid-β and mitochondria in aging and Alzheimer’s disease: implications for synaptic damage and cognitive decline. Journal of Alzheimer’s disease 20, S499–S512. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Reddy PH, Tripathi R, Troung Q, Tirumala K, Reddy TP, Anekonda V, Shirendeb UP, Calkins MJ, Reddy AP, Mao P, 2012. Abnormal mitochondrial dynamics and synaptic degeneration as early events in Alzheimer’s disease: implications to mitochondria-targeted antioxidant therapeutics. Biochimica et Biophysica Acta (BBA)-Molecular Basis of Disease 1822, 639–649. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rinn JL, Chang HY, 2012. Genome regulation by long noncoding RNAs. Annual review of biochemistry 81, 145–166. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sabin LR, Delás MJ, Hannon GJ, 2013. Dogma derailed: the many influences of RNA on the genome. Molecular cell 49, 783–794. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schonrock N, Humphreys DT, Preiss T, Götz J, 2012. Target gene repression mediated by miRNAs miR-181c and miR-9 both of which are down-regulated by amyloid-β. Journal of Molecular Neuroscience 46, 324–335. [DOI] [PubMed] [Google Scholar]
- Sehar U, Rawat P, Reddy AP, Kopel J, Reddy PH, 2022. Amyloid Beta in Aging and Alzheimer’s Disease. Int J Mol Sci. 23(21), 12924. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sethi P, Lukiw WJ, 2009. Micro-RNA abundance and stability in human brain: specific alterations in Alzheimer’s disease temporal lobe neocortex. Neuroscience letters 459, 100–104. [DOI] [PubMed] [Google Scholar]
- Sheinerman KS, Tsivinsky VG, Abdullah L, Crawford F, Umansky SR, 2013. Plasma microRNA biomarkers for detection of mild cognitive impairment: biomarker validation study. Aging (Albany NY) 5, 925. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shi Y, Massagué J, 2003. Mechanisms of TGF-β signaling from cell membrane to the nucleus. cell 113, 685–700. [DOI] [PubMed] [Google Scholar]
- Shoshan E, Mobley AK, Braeuer RR, Kamiya T, Huang L, Vasquez ME, Salameh A, Lee HJ, Kim SJ, Ivan C, 2015. Reduced adenosine-to-inosine miR-455-5p editing promotes melanoma growth and metastasis. Nature cell biology 17, 311–321. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Swerdlow RH, 2011. Brain aging, Alzheimer’s disease, and mitochondria. Biochimica et Biophysica Acta (BBA)-Molecular Basis of Disease 1812, 1630–1639. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Swingler TE, Niu L, Pontifex MG, Vauzour D, Clark IM, 2022. The microRNA-455 null mouse has memory deficit and increased anxiety, targeting key genes involved in Alzheimer’s disease. International journal of molecular sciences 23, 554. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Swingler TE, Wheeler G, Carmont V, Elliott HR, Barter MJ, Abu-Elmagd M, Donell ST, Boot-Handford RP, Hajihosseini MK, Münsterberg A, 2012. The expression and function of microRNAs in chondrogenesis and osteoarthritis. Arthritis & Rheumatism 64, 1909–1919. [DOI] [PubMed] [Google Scholar]
- Tampellini D, Gouras GK, 2010. Synapses, synaptic activity and intraneuronal Aβ in Alzheimer’s disease. Frontiers in aging neuroscience 2, 1330. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Terry RD, Masliah E, Salmon DP, Butters N, DeTeresa R, Hill R, Hansen LA, Katzman R, 1991. Physical basis of cognitive alterations in Alzheimer’s disease: synapse loss is the major correlate of cognitive impairment. Annals of Neurology: Official Journal of the American Neurological Association and the Child Neurology Society 30, 572–580. [DOI] [PubMed] [Google Scholar]
- Tesseur I, Zou K, Esposito L, Bard F, Berber E, Van Can J, Lin AH, Crews L, Tremblay P, Mathews P, 2006. Deficiency in neuronal TGF-β signaling promotes neurodegeneration and Alzheimer’s pathology. The Journal of clinical investigation 116, 3060–3069. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Turchinovich A, Samatov TR, Tonevitsky AG, Burwinkel B, 2013. Circulating miRNAs: cell–cell communication function? Frontiers in genetics 4, 53855. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ueberham U, Ueberham E, Gruschka H, Arendt T, 2006. Altered subcellular location of phosphorylated Smads in Alzheimer’s disease. European Journal of neuroscience 24, 2327–2334. [DOI] [PubMed] [Google Scholar]
- Van den Hove DL, Kompotis K, Lardenoije R, Kenis G, Mill J, Steinbusch HW, Lesch K-P, Fitzsimons CP, De Strooper B, Rutten BP, 2014. Epigenetically regulated microRNAs in Alzheimer’s disease. Neurobiology of aging 35, 731–745. [DOI] [PubMed] [Google Scholar]
- Vilardo E, Barbato C, Ciotti M, Cogoni C, Ruberti F, 2010. MicroRNA-101 regulates amyloid precursor protein expression in hippocampal neurons. Journal of Biological Chemistry 285, 18344–18351. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Voinnet O, 2009. Origin, biogenesis, and activity of plant microRNAs. Cell 136, 669–687. [DOI] [PubMed] [Google Scholar]
- Wang H, Liu J, Zong Y, Xu Y, Deng W, Zhu H, Liu Y, Ma C, Huang L, Zhang L, 2010. miR-106b aberrantly expressed in a double transgenic mouse model for Alzheimer’s disease targets TGF-β type II receptor. Brain research 1357, 166–174. [DOI] [PubMed] [Google Scholar]
- Wang J, Yang S, Li W, Zhao M, Li K, 2022. Circ_0000491 promotes apoptosis, inflammation, oxidative stress, and fibrosis in high glucose-induced mesangial cells by regulating miR-455-3p/Hmgb1 Axis. Nephron 146, 72–83. [DOI] [PubMed] [Google Scholar]
- Wang Y, Shen Y, 2022. Exosomal miR-455-3p from BMMSCs prevents cardiac ischemia-reperfusion injury. Human & experimental toxicology 41, 09603271221102508. [DOI] [PubMed] [Google Scholar]
- Wen X, Li H, Sun H, Zeng A, Lin R, Zhao J, Zhang Z, 2020. miR-455-3p reduces apoptosis and alleviates degeneration of chondrocyte through regulating PI3K/AKT pathway. Life sciences 253, 117718. [DOI] [PubMed] [Google Scholar]
- Winblad B, Palmer K, Kivipelto M, Jelic V, Fratiglioni L, Wahlund LO, Nordberg A, Bäckman L, Albert M, Almkvist O, 2004. Mild cognitive impairment–beyond controversies, towards a consensus: report of the International Working Group on Mild Cognitive Impairment. Journal of internal medicine 256, 240–246. [DOI] [PubMed] [Google Scholar]
- Wu L, Belasco JG, 2008. Let me count the ways: mechanisms of gene regulation by miRNAs and siRNAs. Molecular cell 29, 1–7. [DOI] [PubMed] [Google Scholar]
- Yamada Y, Arai T, Kojima S, Sugawara S, Kato M, Okato A, Yamazaki K, Naya Y, Ichikawa T, Seki N, 2018. Anti-tumor roles of both strands of the miR-455 duplex: their targets SKA1 and SKA3 are involved in the pathogenesis of renal cell carcinoma. Oncotarget 9, 26638. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yao S, Tang B, Li G, Fan R, Cao F, 2016. miR-455 inhibits neuronal cell death by targeting TRAF3 in cerebral ischemic stroke. Neuropsychiatric disease and treatment, 3083–3092. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yun CW, Lee JH, Lee SH, 2019. Hypoxia-induced PGC-1α regulates mitochondrial function and tumorigenesis of colorectal cancer cells. Anticancer research 39, 4865–4876. [DOI] [PubMed] [Google Scholar]
- Zeng Y, Gao T, Huang W, Yang Y, Qiu R, Hou Y, Yu W, Leng S, Feng D, Liu W, 2019. MicroRNA-455-3p mediates GATA3 tumor suppression in mammary epithelial cells by inhibiting TGF-β signaling. Journal of biological chemistry 294, 15808–15825. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhan T, Huang X, Tian X, Chen X, Ding Y, Luo H, Zhang Y, 2018. Downregulation of MicroRNA-455-3p links to proliferation and drug resistance of pancreatic cancer cells via targeting TAZ. Molecular Therapy-Nucleic Acids 10, 215–226. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang H, Guan M, Townsend KL, Huang TL, An D, Yan X, Xue R, Schulz TJ, Winnay J, Mori M, 2015. Micro RNA-455 regulates brown adipogenesis via a novel HIF 1an-AMPK-PGC 1α signaling network. EMBO reports 16, 1378–1393. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang S, Wu W, Jiao G, Li C, Liu H, 2018. miR-455-3p activates Nrf2/ARE signaling via HDAC2 and protects osteoblasts from oxidative stress. Int J Biol Macromol 107, 2094–2101. [DOI] [PubMed] [Google Scholar]
- Zhang Y, Fan K, Xu X, Wang A, 2020. The TGF-β1 induces the endothelial-to-mesenchymal transition via the UCA1/miR-455/ZEB1 regulatory axis in human umbilical vein endothelial cells. DNA and cell biology 39, 1264–1273. [DOI] [PubMed] [Google Scholar]
- Zhao Y, Jaber VR, LeBeauf A, Sharfman NM, Lukiw WJ, 2019. microRNA-34a (miRNA-34a) mediated down-regulation of the post-synaptic cytoskeletal element SHANK3 in sporadic Alzheimer’s disease (AD). Frontiers in Neurology 10, 28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zheng J, Lin Z, Zhang L, Chen H, 2016. MicroRNA-455-3p inhibits tumor cell proliferation and induces apoptosis in HCT116 human colon cancer cells. Medical Science Monitor: International Medical Journal of Experimental and Clinical Research 22, 4431. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhu X, Li H, Tian M, Zhou S, He Y, Zhou M, 2020. miR-455-3p alleviates propofol-induced neurotoxicity by reducing EphA4 expression in developing neurons. Biomarkers 25, 685–692. [DOI] [PubMed] [Google Scholar]
- Zovoilis A, Agbemenyah HY, Agis-Balboa RC, Stilling RM, Edbauer D, Rao P, Farinelli L, Delalle I, Schmitt A, Falkai P, 2011. microRNA-34c is a novel target to treat dementias. The EMBO journal 30, 4299–4308. [DOI] [PMC free article] [PubMed] [Google Scholar]
