Abstract
The physicochemical characteristics of RNA admit non-coding RNAs to perform a different range of biological acts through various mechanisms and are involved in regulating a diversity of fundamental processes. Notably, some reports of pathological conditions have proved abnormal expression of many non-coding RNAs guides the ailment. Vault RNAs are a class of non-coding RNAs containing stem regions or loops with well-conserved sequence patterns that play a fundamental role in the function of vault particles through RNA-ligand, RNA–RNA, or RNA-protein interactions. Taken together, vault RNAs have been proposed to be involved in a variety of functions such as cell proliferation, nucleocytoplasmic transport, intracellular detoxification processes, multidrug resistance, apoptosis, and autophagy, and serve as microRNA precursors and signaling pathways. Despite decades of investigations devoted, the biological function of the vault particle or the vault RNAs is not yet completely cleared. In this review, the current scientific assertions of the vital vault RNAs functions were discussed.
Keywords: ncRNA, Vault complex, Vault RNA, vtRNA, viral infection, tumorigenesis
Introduction
Recently, the importance of diverse and surprising types of non-protein coding RNA (ncRNA) molecules have been widely recognized. In addition, all transcripts analyzed to date consist of two different classes of RNA molecules, mRNAs, which are generally translated into proteins by a series of ribosomes, and ncRNAs, which carry out cellular functions at the RNA level.1 In general, the existence of ncRNAs has been observed in all three kingdoms (Archaea, Bacteria, and Eukarya) and their number increases as organisms become more complex.1,2 Deep sequencing techniques have enabled a complete understanding of cellular transcriptomes, and it has been shown that primitive single-celled eukaryotes (e.g., yeast) or prokaryotes contain fewer ncRNA genes than multicellular eukaryotes. Additionally, it has been estimated mammals encode around 100,000 ncRNAs. However, this knowledge of ncRNA is essential for understanding the complexity of mammals compared to “lower organisms” despite their slightly greater number of protein-coding genes.2, 3, 4, 5, 6
Some data show approximately 97%–98% of the transcriptional output of the human genome is ncRNA.7, 8, 9, 10 Over the last few years, ncRNAs have attracted a lot of attention and can divide into common ncRNAs like rRNA, tRNA, small nuclear RNA, small nucleolar RNA, etc. New types of ncRNAs include microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNA (circRNA).11, 12, 13 Furthermore, human cells express several cytoplasmic ncRNAs, including vault RNAs (vtRNAs) and brain cytoplasmic RNAs (BC RNAs,14 Y RNA,15 etc.). Interestingly, the physicochemical properties of RNA enable ncRNAs to perform a diversity of cellular functions through different mechanisms.1,16 They are involved in the regulation of many basic processes, including transcription, translation, RNA processing, mRNA turnover, DNA replication, genomics stability, gene expression, chromatin remodeling, and protein stability and localization.1,16, 17, 18, 19, 20 It should be noted that deregulated expression of ncRNA is a major cause of various pathological conditions.21
Vault complex
Vault components and structure
Interestingly, early findings revealed a highly conserved subset of ncRNAs derived from vault RNAs, so named because they were first discovered as part of the ribonucleoprotein complex known as the vault. Although they were first described 30 years ago in a Rome laboratory in 1986, vault RNAs are largely unknown and their molecular role is still under investigation.21,22 Vaults are revealed in partially purified fractions of human fibroblasts, mouse 3T3 cells, glial cells, and rabbit alveolar macrophages. These new ribonucleoprotein structures appear to be common in different cell types.21,22
The vaults are large cytoplasmic ribonucleoprotein particles with a sedimentation value of approximately 150 S.23,24 Based on the nature of the structure, the particle size of the vault was determined to be ∼55 × 30 nm, with a molecular mass of approximately 13 MDa (three times the size of a ribosome). The intact vault particle has a double symmetry, where each half-vault can begin in the shape of a flower with eight petals around a central ring. The remarkable conservation and wide distribution of the vaults suggest that their functions are essential for eukaryotic organisms and the structure of the particles must be important for their functions.25, 26, 27, 28
Moreover, the vaults are mainly located in the cytoplasm and can classify as cytoskeletal elements. In addition, part of the vault has been located on the cytoplasmic side of the nuclear membrane within or near the nuclear pore complex.25,29, 30, 31, 32, 33 However, their different morphology and subcellular distribution suggest an intracellular role34, 35, 36 and nucleocytoplasmic transport processes.25,31,33 The stoichiometric model of the vault particle suggests that it contains six copies of vtRNA, but these represent 5% of the total mass of the dome complex. Interestingly, 95% of vtRNAs are evenly distributed in the cytoplasm, while the remaining only 5% are directly associated with vault particles.30,37,38 It is important to point out that the vaults have a highly conserved morphology in eukaryotes and are widespread in eukaryotes.27 Vault RNA is an integral part of the large vault particle, a hollow barrel-shaped ribonucleoprotein (RNP) complex composed of multiple copies of three proteins: major vault protein (MVP), poly (ADP-ribose) vault polymerase (vPARP), and telomerase-associated protein 1 (TEP1). MVP is the main structural protein of the vault complex, accounting for about 70% of the mass of the particles, and self-assembles in vivo to form vault-shaped particles.28,39 It was recently shown that the absence of TEP1 completely disrupts the stable binding of vtRNA to purified vault particles and reduces the concentration and stability of the vault RNA. Thus, a novel role for TEP1 in vivo was discovered as an integral vault protein important for the stabilization and recruitment of the vault RNA into the vault particle.10,40 Furthermore, the precise role of this subunit in the telomerase complex is still unclear, except it is capable of binding telomerase RNA.10
Genomic organization of multiple vault RNA genes and homologous evolution
Two vaults RNA loci are syntonically conserved in most mammals,41 and varying numbers of paralogs across animal kingdoms have been reported.41,42 Mechanistically, four human vtRNA paralogs have been identified and three related human vtRNA genes (hvg) have been described (hvg1-3; GenBank™ accession numbers: AF045143, AF045144, and AF045145).10,30 Human vtRNA gene sequences revealed that the vtRNA-1 locus located between the zinc-finger matrin-type 2 (ZMAT2) and the proto-cadherin cluster (PCHD) gene, which carries the genetic information for three hvg1-3 vault RNAs (vtRNA1-1, vtRNA1-2, vtRNA1-3) that are found in triplicate position 5q31.3 chromosome. The vtRNA-2 is a locus located between the genes encoding transforming growth factor beta 1 (TGFB1) and SMAD family member 5 (SMAD5) on chromosome 5q31.3, which encodes vtRNA2-1 (hvg-5).30,37,43 In addition, in the hg38 human genome assembly, two vtRNA pseudogenes, vtRNA2-2p and vtRNA3-1p (formerly hvg-4) were each annotated on chromosomes 2p14 and Xp11.2, respectively.37,41,43 Experimental evidence also suggests miR-886 is a vtRNA and not a canonical miRNA, as sequence alignment clearly identifies this sequence as either a vtRNA homolog or a vault complex associated molecule (later a homolog divergent of vtRNA2-1).37,41 Therefore, the revised name “vtRNA2-1” was approved by the HUGO Genetic Nomenclature Committee.44,45 Human vtRNA (hvtRNA) length ranges from 88 to 150 nt, with hvg-1 encoding 98-nt RNAs and hvg-2 and hvg-3 encoding similar 88-nt RNAs. All three genes are located in the 16 kb regions of chromosome 5, suggesting that these genes arose in tandem gene duplication.30,32 Nevertheless, hvg-4 seems to be unexpressed32 (Fig. 1).
Figure 1.
Overview of the evolution and location of vtRNAs genes. (A) In most mammals, the two syntenically vtRNA loci are highly conserved. Of the four human paralogs of vtRNA, hvg1-3 are located on the 5q31.3 chromosome between the ZMAT2 and the PCHD gene. vtRNA2-1 locus is located between the TGFB1 and SMAD5 gene on chromosome 5q31.3. Furthermore, two vtRNA pseudogenes, vtRNA2-2p and vtRNA3-1p were annotated on chromosomes 2p14 and Xp11.2, respectively. Human vtRNAs are 88–150 nt in length, which are transcribed by RNA polymerase III and contain standard internal promoter elements of A and B. Also, most mammals, including mice and rats, have a single copy of the vtRNA gene on the PCDH locus. (B) The extended structure of vtRNAs of chimpanzees, mice, and rats. The positions of the internal transcription elements of boxes A and B are indicated, and the conserved sequences are highlighted.
The four genes transcribed by RNA polymerase III (Pol III) type 2 contain standard internal promoter elements of the A and B boxes with various external promoter elements and have a secondary core. Although the ring chain structure is well conserved,37,41 only the hvg1-3 gene contains external type-3 TATA, proximal, and distal sequence elements. In addition, it has been reported the sequences between A and B boxes are unique and affect the transcription efficiency. The sequence length among these elements is 41, 31, 31, and 44 bases in hvg-1, hvg-2, hvg-3, and hvg-4, respectively.46 Interestingly, vtRNAs isolated from different species have similar predicted secondary structures, despite differences in length, suggesting the association of vtRNAs with reservoirs is not random.47 The vault RNA sequence of a human, mouse, rat, and bullfrog was determined.23,30,32 Since vault RNA genes are only available for a few organisms, it is initially impossible to conclude whether there was a single gene or multiple copies.23,46
The human vtRNA-1 locus encodes three paralogues and their orthologs are also found in the phylogenetically close chimp (P. troglodytes). Instead, only two vtRNA-1 orthologs are encoded in the genome of other primates. This clue and sequence alignment analysis revealed that vtRNA1-2 and vtRNA1-3 are the result of recent gene duplication. Most mammals, including rats and related mouse models (M. musculus), have a single copy of vtRNA-1 at the PCDH locus.41 Significantly, rat vtRNA is 144 bases in length while bullfrog vtRNA exists as two highly related species of 89 and 94 bases. In addition, the rat vtRNA gene contains two B-box consensus elements separated by 34 bp. Rat and bullfrog vtRNAs contain sequences that correspond to the internal promoter elements of genes transcribed by RNA polymerase III.23,24
Particularly, in most mammals, the vtRNA-2 locus lacks or encodes only one paralog vtRNA, exception for the sloth genome (C. hoffmanni) which contains two paralogs, vtRNA-2 48. Furthermore, the existence of vtRNA has been shown in the sea urchin (Strongylocentrotus purpuratus). However, the sequences were not determined.49 The vtRNA locus has been sequenced in other classes, including two copies of vtRNAs from R. catesbeiana,23 four paralogues in H. sapiens,10 and one each in S. purpuratus,49 T. brucei,50 D. rerio and O. latipes.41 In addition, the development of an iterative algorithm identified more than 100 potential vtRNA genes in the deuterostome genome.41
Interestingly, most mammals have a single vtRNA copy at the PCDH locus. In a few cases, notably in primates, guinea pig (Cavia porcellus), pika (Ochotona princeps), and shrew (Sorex araneus), they grow in a locally multicopy cluster.41 Besides, the dog (Canis familiaris) genome contains only a degraded pseudogene of vtRNA at the PCDH locus. Instead, there is a single alternative sequence (antisense direction) between TGFB1 and SMAD5.37,41 Similarly, in gnathostomes, the functional vtRNA-1 gene is often located upstream of the PCDH cluster. However, in the Eutheria, there is a second locus with a functional vtRNA-2 gene between the TGFB1 and SMAD5 genes. In several eutherian lineages, only one of the two loci contains the vtRNA gene. This apparent complementation strongly supports the conclusion that the transcript of the vtRNA-2 locus is indeed an additional vtRNA.37 There are clear differences between vtRNA1-1 versus vtRNA1-2 and vtRNA1-3 sequences only within primates, suggesting that gene duplication occurred early in the primate lineage. Since the molecular function of vtRNA is still unknown, we cannot speculate on what might be the reason for this lineage-specific evolutionary pattern.41
Vault RNAs functions
More recently, only a small fraction of vtRNAs is associated with the vault complexes,30,51 suggesting that the cellular function of ncRNAs is outside of the vault complex.52 The ends (5′ and 3′) of vtRNA form a stem-like structure, but the internal sequences of these RNAs are folded differently. These predictions indicate that vtRNA plays an important role in vault particle function, probably through RNA, RNA–RNA, or RNA-protein ligand interactions.53,54 Taken together, vtRNAs have been proposed to be involved in a variety of functions, including cell proliferation,55,56 nucleocytoplasmic transport, intracellular detoxification processes, and therefore a role in multidrug resistance (MDR),30,53,57, 58, 59, 60, 61, 62 signaling pathway,63,64 apoptosis,65,66 innate immune response,67 autophagy,68 which serve as miRNA precursors.69 Many of these results are consistent with the hypothesis that altered vtRNA expression is associated with tumorigenesis,66,70 viral defense,52 and modulation of the metabolism of different classes of RNA.50 Although the functional role of F. hepatica vtRNA is unknown, their selective packaging in the fluke extracellular vehicles (EVs) implies they may be involved in host–parasite interactions.71 Despite decades of dedicated research, the biological function of the vault particle or vtRNA has not yet been fully elucidated. It is evident that vtRNAs are vital regulatory molecules that orchestrate intracellular functions and can either drive or prevent pathophysiological processes (Table 1). The presence of this molecule in extracellular RNA circulating in liquid biopsies, serum,72,73 EVs from human samples, axons, and neurons,74,75 and shuttle RNA,76 makes it a promising candidate biomarker for the diagnosis and prediction of various diseases. This review summarizes the known functions of vtRNAs and their involvement in cellular mechanisms (Fig. 2).
Table 1.
A summary of validated dysregulated vault RNAs in different cellular processes.
| ncRNA | Patients and cells tested | Related process | Description | Reference |
|---|---|---|---|---|
| vtRNA1-1 | Animal models (mouse, rabbit, guinea pig) and cortical neuron culture | Synapse formation | hvtRNA1-1 increases the expression of synaptic marker proteins, ERKs phosphorylation, and the number of PSD-95 and synapsin I, thus modifying the modulation of the MAPK signaling pathway can improve synapse formation. | 95,96 |
| Huh-7 cell line, murine cells | Autophagy | vtRNA1-1 contributes to the regulation of the autophagic flux, by directly interacting with the autophagy receptor protein SQSTM1/p62. | 68,137 | |
| EBV-infected human B cells | Viral infection | Ectopic expression of vtRNA1-1 leads to increased viral establishment and reduced apoptosis in B cells, leaving cells vulnerable to efficient EBV infection. | 66 | |
| HeLa cell line | Apoptotic signaling | The PI3K/Akt pathway and the ERK1/2-MAPK cascade are dysregulated in the vtRNA-1 knockout cell lines, thus eliciting increased levels of apoptosis in prolonged starvation. | 70 | |
| SCs skin | Cellular differentiation | Methylated vtRNA1-1 prevents the binding of SRSF2 and NSUN2 to its putative binding site spanning methylated C69, which in turn inhibits epidermal differentiation. | 155,158 | |
| HCC | Multidrug resistance | vtRNA1-1 knockdown in HCC cells leads to lysosomal compartment dysfunction by inhibiting TFEB nuclear translocation and increasing activation of the MAPK, resulting in lysosome-mediated resistance to chemotherapy. | 137 | |
| BCC line | Multidrug resistance and cell proliferation | vtRNA1-1 releases the PSF protein, allowing the GAGE6 oncogene to become activated and thereby inducing drug resistance by promoting cell proliferation. | 56 | |
| vtRNA1-3 | MDS | Cell proliferation | vtRNA1-3 promoter hypermethylation is associated with a decreased overall survival of MDS. | 87 |
| vtRNA2-1 (nc886) | AML | Cell proliferation | vtRNA2-1 promoter hypermethylation in AML induces pPKR down-regulation, which strikingly has a significantly poorer outcome in AML patients. | 86 |
| GC | Cell proliferation | nc886 inhibits cell proliferation in GC cells via suppression of oncogenes such as FOS, NF-κB, and MYC. | 55 | |
| PCa | Cell proliferation | Hypermethylation of nc886 promoter in tumor tissue correlates with clinical staging of PCa, including biochemical recurrence, clinical T-value, and Gleason scores through increased cell proliferation and invasion. | 44,179 | |
| HCC | Cell proliferation | Hypermethylation of the vtRNA2-1 promoter is increased in stage III HCC tumors compared with stage I & II tumors and is also significantly associated with HCC tumor recurrence. | 90 | |
| PD | Cell proliferation | Hypermethylation of 15 CpG sites in the vtRNA2-1 gene can be associated with PD. | 91 | |
| T2DM | Glycemic response | Hypomethylation of vtRNA2-1 becomes considerably related to a poor glycemic reaction to GLP-1 treatment for T2DM. | 98 | |
| IgA nephropathy | Cell proliferation | Hypermethylation of vtRNA2-1 in IgAN patients led to a decreased CD4+ T-cell proliferation following TCR stimulation and to the overexpression of TGFβ. | 100 | |
| HaCaT cells | Sensitivity to UVB | nc886 expression decreases by UVB radiation, while PKR phosphorylation thru MAPKs will increase, and will increase the expression of inflammatory cytokines, MMP-9, and COX-2, which in the end boost inflammatory responses. | 105 | |
| SLE | Cell proliferation | Overexpression of vtRNA2-1 on B cells is associated with SLE through specific binding to PKR. | 180 | |
| Pregnant women | Preterm birth | Hypermethylation of the vtRNA2-1 promoter results in an increased risk of PTB caused by the pro-inflammatory cytokines. | 181 | |
| RCC | Apoptotic signaling | Overexpression of nc886 promotes A-498 cell proliferation and invasion and inhibits cell apoptosis by activating JAK2/STAT3 signaling. | 151 | |
| CCC | Multidrug resistance | E2F1 promotes nc886 transcription and turns MVP expression sufficiently to drive drug resistance in cervical cancer cells. | 174 | |
| svtRNA2-1a | PD | Cell proliferation | Up-regulation of svtRNA2-1a in PD contributes to the disruption of gene expression networks, underlying metabolic impairments, and cellular dysfunction. | 94 |
| vtRNA2-1-5p | CSCC | Apoptotic signaling | Overexpression of vtRNA2-1-5p in human CSCC increases cervical cancer cell invasion, proliferation, and tumorigenicity while decreasing apoptosis and p53 expression. | 150 |
| svRNAb | HepG2 cells | Multidrug resistance | svRNAb down-regulates CYP3A4, a key enzyme in drug metabolism, causing drug resistance. | 69,171 |
Abbreviations: AKT, protein kinase B; AML, acute myeloid leukemia; BCC, breast cancer cell line; CCC, cervical cancer cell; COX-2, cyclooxygenase 2; CSCC, cervical squamous cell carcinoma; CYP3A4, cytochrome P450 3A4; EBV: Epstein–Barr virus; ERKs, extracellular signal-regulated kinases; GAGE6, G antigen 6; GC, gastric cancer; HCC, hepatocellular carcinoma; Hela cell line, Henrietta Lacks cell line; JAK2/STAT3, Janus kinase 2 signal transducer and activator of transcription 3; MAPK, mitogen-activated protein kinase; MDS, myelodysplastic syndrome; ncRNA, non-coding RNA; NF-Κb, nuclear factor kappa light chain enhancer of activated B cells; NSUN2, NOP2/Sun RNA methyltransferase 2; PCa, prostate cancer; PD, Parkinson's disease; PI3K, phosphatidylinositol 3-kinase; PKR, protein kinase R; PSD-95, postsynaptic density protein 95; PSF, PTB-associated splicing factor/splicing factor proline–glutamine rich (PSF or SFPQ); PTB, preterm birth; RCC, renal cell carcinoma; SCs, stem cells; SQSTM1, Sequestosome 1; SRSF2, serine/arginine-rich splicing factor 2; SLE, systemic lupus erythematosus; TFEB, transcription factor EB; TGF β, transforming growth factor β; T2DM, type 2 diabetes mellitus; vtRNA, vault RNA.
Figure 2.
Possible mechanisms of vault RNAs are involved in different cellular processes and contribute to viral infections and tumorigenesis. Vault RNAs are a class of ncRNAs that contain stem-loop regions with well-conserved sequence patterns. They consist of only four members in humans and play fundamental roles in cells, possibly through RNA-ligand, RNA–RNA, or RNA-protein interactions. Accordingly, it has been proposed that vault RNAs are involved in cell proliferation and modulate the metabolism of multiple RNA classes: intracellular transfer, nucleocytoplasmic transport, regulation of central signaling pathways, regulation of cell communication, and more recently, nuclear pore complex formation. Furthermore, vtRNAs play an important role in the immune response, apoptosis, autophagy, detoxification processes, and multidrug resistance mechanisms. Most of our current knowledge of the functions of vtRNAs focuses on viral infections and cancer.
Vault RNAs and cancer
Cancer is an epigenetic and genetic disorder. However, the interaction between these two processes is unclear.77, 78, 79 Recent advances in genomic technology have identified thousands of ncRNAs in the human genome. Although the role of ncRNAs in cancer epigenetics has been demonstrated, most have not been assigned a role.80,81 ncRNAs have emerged as key players in cancer initiation and progression,82, 83, 84 therefore, their clinical value is the subject of intense investigation.44,85 An interesting observation regarding the role of vtRNA is the association of vtRNA with the vault complex in several human cancer cell lines.32,53,66,70
DNA methylation status of vault RNAs
Hematopoietic diseases
Acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS) belong to a heterogeneous group of clonal hematopoietic diseases. MDS and AML are often associated with deletions at chromosome 5q and aberrant DNA methylation patterns, including hypermethylation, indicating the presence of important tumor suppressors at this locus.86,87 In normal hematopoiesis, vtRNA shows differential methylation between different hematopoietic cell populations, suggesting that allele-specific methylation may occur during hematopoiesis.87 Treppendahl and colleagues demonstrated that AML patients with deficient vtRNA2-1 methylation had significantly bettered outcomes than patients with mono or biallelic methylation. In other words, this probably implies that a higher gene dose influences the outcomes. Consistent with the result of this study, the methylation is inversely proportional to vtRNA2-1 expression, and 5-azanucleosides induce vtRNA2-1 and reduce phosphorylated RNA-dependent protein kinase (pPKR) whose activity is regulated by vtRNA2-1. Since pPKR promotes cell survival in AML, the data are consistent with vtRNA2-1 as a tumor suppressor in AML.86 In addition, vtRNA1-3 promoter hypermethylation is frequent in lower-risk MDS patients and is associated with a decreased overall survival.87
Gastric cancer
Lee et al investigated that nc886 suppresses cell proliferation when ectopically expressed in gastric cancer cells by suppressing oncogenes such as FOS, NF-κB, and MYC, so nc886 CpG hypermethylation and corresponding down-regulation are significantly associated with lower survival rates in gastric cancer patients.55 The nc886 RNA is a confirmed tumor suppressor ncRNA that acts by down-regulating the cancer survival factor pPKR.86,88 The pre-miR-886 was further found to be physically associated with PKR, an interferon-inducible and double-stranded RNA-dependent kinase.89 Deletion of pre-miR-886 reduces cell proliferation through activation of PKR and its downstream pathways, phosphorylation of eIF2α, and the NF-κB pathway. Pre-miR-886 was deleted in some cancer cell lines and clinical samples.89 However, valid data based on the precise identification of nc886 for its functional cellular function as a tumor suppressor has not yet been provided.
Hepatocellular carcinoma
Similarly, hepatocellular carcinoma (HCC) patients had worse outcomes when the vtRNA2-1 promoter was differentially hypermethylated in tumor tissues compared with adjacent normal tissues. In addition, vtRNA2-1 promoter methylation is increased in stage III HCC compared with stages I and II. These results indicate that differential hypermethylation of the vtRNA2-1 promoter is significantly associated with tumor recurrence.90
Based on the above observations regarding the involvement of vtRNA in the pathology of various tumors, it can be concluded that vtRNAs are proposed as a new type of ncRNA that acts as a tumor suppressor (TSG) gene which inhibits PKR and vice versa. In contrast, some reports have shown that vtRNAs act as oncogenes in tumor progression, development, and invasion. Therefore, it is a potential prognostic biomarker and therapeutic target for tumors.
Vault RNAs and non-communicable diseases
Age-related neurodegenerative disease
Parkinson's disease (PD) is known to be the second most common age-related neurodegenerative disease. Nowadays, it is most accurately diagnosed in the advanced stage with a series of motor deficits preceded by a litany of non-motor symptoms that appear over years or decades.91,92 Notably, recent studies found that blood-based gene expression or epigenetic biomarkers exist across a range of PD diseases and are highly desirable and non-invasively detectable markers.91,93
Recently, Henderson et al reported hypermethylation of 15 CpG sites encompassing the vtRNA2-1 gene in the whole blood of Parkinson's disease.91 A previous study by Miñones-Moyano et al also showed that a small non-coding RNA considered as a fragment of vtRNA2-1 (svtRNA2-1a) was up-regulated in the amygdala and frontal cortex of patients with advanced PD. Moreover, a double up-regulation was also found in preclinical cases of Alzheimer's disease. The expression patterns of svtRNA2-1a during human brain development and aging and the deleterious results of mimicking svtRNA2-1a overexpression in neurons further suggest that low levels of svtRNA2-1a may be important for neuronal maintenance. These results proposed that early dysregulation of svtRNA2-1a in PD may lead to disruption of gene expression networks, metabolic disturbances, and cellular dysfunction.94
Synapse formation
Recent evidence has been provided for a novel role for vtRNA in synapse formation. New studies show that mouse vtRNA (mvtRNA) promotes synapse formation by regulating mitogen-activated protein kinase (MAPK) signaling. mvtRNA binds to and activates mitogen-activated protein kinase 1 (MEK1) in dendrites, enhancing MEK1-mediated extracellular signal-regulated kinase activation, thereby regulating synapse generation.95 In addition, the expression of hvtRNA1-1, but not hvtRNA2-1, increases the expression of synaptic marker proteins, extracellular signal-regulated kinase (ERK) phosphorylation, and the number of double-positive postsynaptic density protein 95 (PSD-95), and synapsin I to a similar extent as mvtRNA. According to this knowledge, hvtRNA1-1 modifies the modulation of the MAPK signaling pathway to improve synapse formation.96 Therefore, a detailed understanding of the mechanisms of synapse formation will allow further elucidation of the pathogenesis of neurodevelopmental disorders. Furthermore, a better understanding of the signaling complexes involved in vault complexes could lead to the development of new therapeutic strategies for neurological disorders.95
Type 2 diabetes
Glucagon-like peptide-1 (GLP-1) is an incretin that plays a critical physiological role in glucose homeostasis, which can stimulate the secretion of insulin produced by the intestine during meals and keeps the pancreatic β-cells healthy and proliferating.97 The therapeutic efficacy of GLP-1 analogs in type 2 diabetes (T2DM) is 50%–70%. In support of this, the discovery of genetic biomarkers is still needed to predict the therapeutic efficacy of GLP-1 analogs for treatment. Today, the glycemic response to GLP-1 analog therapy is associated with the methylation status of the vtRNA2-1 promoter and rs2346018 polymorphism. Furthermore, hypomethylation of vtRNA2-1 (<40% methylation) is significantly associated with a poor glycemic response to GLP-1 treatment.98
Glomerulonephritis
IgAN (IgA nephropathy) is the most common form of primary glomerulonephritis worldwide and has a strong genetic component.99,100 In this case, DNA methylation may also be an important factor influencing the disease.100,101 Sallustio et al. (2016) identified for the first-time specific regions of DNA that were abnormally methylated in IgAN patients, leading to reduced TCR signaling strength of CD4+ T cells and their abnormal response and activation, resulting in cellular imbalance. In addition, hypermethylation of miR-886 precursors reduced CD4+ T cell proliferation and led to overexpression of transforming growth factor (TGFβ) upon TCR stimulation. This study reveals a novel molecular mechanism underlying the aberrant CD4+ T cell response in IgAN patients.100
Sensitivity to ultraviolet B
Unprotected advertising to ultraviolet (UV) A and B can damage the DNA of pores and skin cells, causing genetic defects or mutations that can result in pores, skin cancer, and various other pores and environmental skin conditions (including premature aging). UV rays can also harm your eyes, including cataracts and eyelid pores, skin cancer, and a variety of environmental pores and skin conditions.102, 103, 104 Under changes in pores and skin cells induced by UVB radiation, down-regulation of nc886 results in depleted phosphorylation of PKR by MAPKs and increased expression of proinflammatory cytokines, matrix metalloproteinase-9 (MMP-9), type IV collagenase, and cyclooxygenase (COX-2), which in the end boost up inflammatory responses and pores and skin aging.105
To sum up, these observations highlight the important role of vtRNAs in the progression of non-infectious diseases through various mechanisms such as PKR-specific binding and signaling pathways, and this mechanism opens new clinical perspectives for the treatment of non-infectious diseases.
Vault RNAs and viral infection
In human viral diseases, infection of host cells is a tightly controlled process, with a temporally regulated expression of viral and cellular genes.106,107 Recently, viral infection has been shown to alter RNA expression profiles at the ncRNA level.37,108 For example, the most up-regulated ncRNAs in the presence of Epstein–Barr virus (EBV) were three host-encoded vtRNAs.22 vtRNA promoted viral replication by inhibiting PKR activation and subsequent responses to antiviral interferon. In addition, increased vtRNA expression was required for efficient inhibition of PKR by NS1 during IAV infection. Viral activation of host vtRNA serves as a mechanism supporting inhibition of PKR signaling, representing a viral strategy to evade host innate immunity.52,109 It is therefore tempting to speculate that the vault complex may be an evolutionary remnant of a primitive viral symbiont of eukaryotic cells.37 Evidence from previous studies suggests the viral infection induces vault RNA expression43,110 in in-vitro models for several viral families, including γ-herpesviridae (herpes simplex virus 1), paramyxovirus (EBV), and influenza-A virus.37,43,52 Therefore, the higher expression of vault RNAs can cause a higher viral load.111
How does vtRNA regulate protein kinase R function?
Protein kinase R (PKR) is an interferon (IFN)- and double-stranded RNA (dsRNA)-activated Ser/Thr protein kinase that is an important part of host innate immunity to viral infection.52,112 Upon activation, PKR phosphorylates its own and downstream substrates, including eukaryotic initiation factor subunit 2α (eIF-2α) and IκB.113,114 Phosphorylated EIF2α dramatically inhibits viral protein synthesis, blocking viral replication.51,113 Furthermore, PKR promotes the activation of nuclear factor kappa B (NF-κB) through phosphorylation of IκB.114 Transcription factor NF-κB positively regulates IFN gene transcription and promotes the expression of IFN-stimulated genes (ISGs).115,116 Influenza A virus (IAV) infection does not activate PKR.117 In contrast, IAV inhibits PKR activity through the virally encoded nonstructural protein (NS1) and the cellular protein p58IPK.118, 119, 120 Previous studies have shown that AVA sequesters and activates p58IPK from heat shock inhibitory protein 40 (Hsp 40), and activated p58IPK inhibits PKR dimerization and phosphorylation through direct interactions between these molecules.121,122 Furthermore, PKR activity can be significantly induced by NS1-deleted virus.123 The viral IAV NS1 protein has been shown to be an inducer that initiates vtRNA up-regulation. The vtRNA silencing animal model showed significantly more resistance to IAV infection, as evidenced by amelioration of acute lung injury and splenic atrophy, thereby increasing survival. Interestingly, vtRNA promoted viral replication by inhibiting PKR activation and subsequent responses to antiviral interferon. In addition, increased vtRNA expression was required for efficient inhibition of PKR by NS1 during IAV infection.123
However, the inhibiting mechanism of PKR activation by NS1 is still unknown. NS1 can cleave PKR dsRNA,118,123 but the affinity between NS1 and dsRNA is low.124 Furthermore, the requirements for the NS1 RNA-binding domain and the direct interaction between NS1 and PKR are controversial based on reports from several research groups.120,125, 126, 127 Of note, in a previous study, nc886 effectively competes with dsRNA for binding to PKR, attenuating dsRNA-mediated PKR activation, thereby suppressing the interferon response. These data suggest that nc886 sets a threshold for PKR activation and triggers activation only when the virus is infected. As discussed above, PKR is activated by various RNA species as well as by actual dsRNA. In the absence of nc886, PKR can be activated by measuring basal levels of this RNA, which can have lethal effects on host cells.128
Vault RNAs and autophagy
Macroautophagy (referred to further as autophagy) is an evolutionarily conserved degradative pathway activated by cellular stress and responsible for the recognition, recycling, elimination, and degradation of intracellular components, organelles, and pathogens in membrane vesicles called autophagosomes.129,130 In this excretory cycle, after engulfing the cargos, the autophagosomes approach and fuse with the lysosomes, breaking down their contents to supply amino acids, lipids, and nucleotides for the anabolic desires of the cells.68 Autophagy removes intracellular substrates ranging from simple ubiquitinated proteins to large proteotoxic aggregates. Human selective autophagy receptor p62 (also known as sequestosome protein 1 (SQSTM1), zeta-interacting protein (ZIP), or orphan receptor co-activator (ORCA)) recruits them into double-membrane vesicles and regulates intracellular load to maintain homeostasis.131, 132, 133, 134 Intracellular pathogenic bacteria and viruses have evolved different defense strategies against autophagy, possibly related to vtRNAs levels.68
Mechanism of vtRNA to modulate autophagy signaling
Recent studies reported the depletion of vtRNA1-1 led to the accumulation of mature autophagosomes, indicating increased autophagic flux as one of the major vtRNAs interacting with p62.68,109 Moreover, up-regulation of vtRNAs levels appears to be a highly effective way to avoid autophagy-targeted viral degradation and subsequent MHC class II antigen presentation.135 Büsche et al. found that small non-coding vtRNA1-1 bind directly to p62 and regulate p62 oligomerization, thus functioning in autophagy.68,136 Similarly, a recent study in Huh-7 cells and mouse cells reported that vtRNA1-1 can help regulate autophagic flow through direct interaction with the autophagy receptor protein SQSTM1/p62 68. Additionally, it was later found the vtRNA1-1 depletion results in the down-regulation of transcription of several catabolism-related genes, including SQSTM1/p62. These data indicate that the reduced expression of SQSTM1/p62 is transcriptionally controlled and does not necessarily depend on the interaction with vtRNA1-1.68,137, 138, 139 This reboregulation affects the aggregate state of p62 and therefore its autophagic degradation and its function as a selective receptor for autophagy.138,139
In addition to its role as an autophagy receptor, p62 provides a platform for important cell signaling pathways.140, 141, 142, 143 It will be interesting to explore worldwide whether the depletion of vtRNA1-1 or the expression of p62 mutants without RNA binding influences other cell signaling pathways.144 The p62/vtRNA1-1 interaction provides a paradigm for the interference of a rib-regulator RNA with protein–protein interactions.144 It has also been shown in detail that the Phox and Bem 1 domain (PB1) and the adjacent linker region (p621–122) are necessary and sufficient for maximal and specific binding of vtRNA1-1. Mutation analysis indicates that K7 and R21 are essential amino acids required for binding to RNA.144 In addition to the previously known role in the oligomerization of p62.145 This result identifies these two residues as cornerstones of riboregulation by which vtRNA1-1 inhibits p62 oligomerization and hence autophagy.144 Furthermore, Ferro et al. showed that vtRNA1-1 knockdown caused alkalinization of intraluminal lysosomal pH, leading to a marked change in lysosome-mediated clearance proteolytic and degradative activity.137
vtRNAs and apoptosis
Apoptosis is a sequential sequence of cell death that occur regularly to ensure a homeostatic balance between the rate of cell formation and cell death. However, dysfunction of this compensatory function can contribute to abnormal cell growth/proliferation or autoimmune diseases, etc.146,147 An increasing number of studies have shown that ncRNAs play an important role in various cellular processes, including proliferation and apoptosis.148,149 For the first time, Amort and colleagues have shown general apoptotic resistance after overexpression of vtRNA1-1 in human B cells infected with Epstein-Barr virus (EBV).66 Here, researchers individually identified latent membrane protein 1 (LMP1) that stimulates vtRNA1-1 expression in BL2 cells as a trigger for NF-κB-dependent up-regulation of vtRNA1-1. Ectopic expression of vtRNA1-1, but not other vtRNA paralogs, results in increased viral establishment and reduced apoptosis, leaving cells vulnerable to efficient EBV infection.66
Another aspect was also revealed, that the knockdown of the main vault protein does not affect these phenotypes, demonstrating that vtRNA1-1, not the vault complex, contributes to a general resistance to cell death, a function located in the central domain of vtRNA1-1. This study describes an NF-KB-mediated role of vtRNA1-1 in inhibiting both extrinsic and intrinsic apoptotic pathways.66 In addition, another group of researchers showed in HeLa knockout cell lines that prolonged starvation caused increased levels of apoptosis in the absence of vtRNA1-1, but not in vtRNA1-3 knockout cells.70 Next-generation deep sequencing of mRNome identified the phosphatidylinositol 3-kinase/protein kinase B (PI3K/Akt) pathway and the ERK1/2-MAPK cascade, of which two critical signaling axes are dysregulated under starvation conditions of mediated cell death in the absence of vtRNA1-1.70 It should be noted that the expression of the vtRNA1-1 mutant identified a short 24-nucleotide stretch of the central domain of vtRNA1-1 as essential for successfully maintaining resistance to apoptosis.70 Similarly, an earlier study from Kong et al. indicates that vtRNA2-1-5p has oncogenic activity linked to cervical cancer progression and the vtRNA2-1-5p also directly targets p53 expression.150 Surprisingly, inhibition of vtRNA2-1-5p has been shown to increase Bcl-2-associated protein X (Bax) expression and apoptotic necrobiosis in cervical cancer cells.150 Inhibition of vtRNA2-1-5p reduced invasion, proliferation, and tumorigenicity of cervical cancer cells while increasing apoptosis and p53 expression.150 These results imply that vtRNA2-1-5p may be a direct regulator of p53. Therefore, it plays a crucial role in the apoptosis and proliferation of cervical neoplastic cells and will be a potential target in the treatment of cervical cancer.150
Similarly, Lei et al. showed that overexpression of nc886 promotes proliferation and invasion of A-498 cells and inhibits cell apoptosis, while deletion of nc886 produces the opposite effects. Additionally, nc886 could activate the Janus kinase 2 signal transducers and the transcription activator 3 (JAK2/STAT3) in A-498 cells. AG490 (tyrphostin), a JAK2 inhibitor, may attenuate the effect of nc886 on cell proliferation, apoptosis, and invasion. This study could represent a useful therapeutic target for renal cell carcinoma (RCC).151
Vault RNAs and cellular differentiation
Stem cells (SC) are unique cells that have an inherent ability to self-renew or differentiate.152,153 These two fateful decisions are tightly regulated at the molecular level by complex signaling pathways. It has been hypothesized the regulation of signaling networks that promote self-renewal or differentiation is largely influenced by the action of transcription factors.152,154 However, small ncRNAs, such as vtRNAs, and their post-transcriptional modifications (epithrescriptome) have emerged as additional regulatory layers that play an essential role in SC fate decisions. Since epitranscriptoma and associated proteome are essential in SCs, they would be considered new tools for the spread of SCs for regenerative medicine.152
The skin is a versatile tissue supported by numerous SCs some of which are unipotent such as epidermal SCs while others like bulge SCs are multipotent.155,156 In NOP2/Sun RNA Methyltransferase 2 (NSUN2) knockout mice, SCs bulging into the hair follicle show delayed differentiation when stimulated during the start of the hair cycle.155 Normally, NSUN2 expression peaks in bulging SCs when they enter anagen, the growth phase of the hair cycle. When NSUN2 ablates in the skin, resting and protruding SCs still accumulate at anagen onset, leading to a general delay in differentiation.155 Similarly, isolated bulge SCs and epidermal SCs from NSUN2 knockout mice show difficulty in in-vitro differentiation.155 Accumulation of 5′tRF disrupts migratory responses in epidermal cells, which may arguably be the underlying mechanism limiting differentiation.155,157 In humans, NSUN2 targets vtRNA1-1 with a single m5C at C69.158,159 Human epidermal progenitors in vitro inhibit differentiation by promoting vtRNA1-1 methylation-dependent processing in small vault RNAs (svRNAs) bound to the RNA-induced silencing complex (RISC).160,161 Methylated vtRNA1-1 prevents the binding of serine/arginine-rich splicing factor 2 (SRSF2) to its purported binding site covering methylated C69. Without the protection conferred by SRSF2 binding, vtRNA1-1 is preferably cleaved into svRNA4, which in turn inhibits epidermal differentiation.160 Exactly how svRNA4 blocks skin differentiation is unclear; however, predicted targets of svRNA4, such as Ovo-like transcriptional repressor 1 (OVOL1), could be its mode of action.161 Indeed, this may be the case, as low levels of OVOL1 are needed to stop epidermal differentiation.160,161
Currently, m5C is the only post-transcriptional RNA modification associated with skin SC functions. Further studies are needed to determine if a different epitranscriptome signature plays a role in skin SCs.152 The presence and absence of RNA modifications regulate RNA metabolism by modulating the binding of writing, reading, and erasing proteins.160,162,163 However, how 5-methylcytosine (m5C) recruits or repels RNA-binding proteins is largely unknown. Methylation of cytosine 69, in vtRNA1-1, occurs frequently in human cells, is mediated exclusively by NSUN2, and determines the processing of vtRNA1-1 into svRNA. Serine/arginine-rich splicing factor 2 (SRSF2) has been identified as a novel vtRNA1-1 binding protein that antagonizes vtRNA1-1 processing by binding the unmethylated form with higher affinity. Both NSUN2 and SRSF2 orchestrate the production of different svRNAs.160 Finally, the research group revealed a direct role for m5C in the processing of vtRNA1-1, including SRSF2, and plays a critical role in the regulation of the epidermal differentiation process.160 Interestingly, identifying the mechanism of molecules involved in the cell differentiation process may be effective for better clinical application of cell differentiation-based therapies in human diseases.
Vault RNAs and drug resistance
Multidrug resistance (MDR) is the most common cause of chemotherapy failure to treat cancer. Multiple mechanisms are responsible for mediating MDR, including overexpression of transmembrane transporters such as ATP binding cassette (ABC) transmembrane transporters such as P-glycoprotein (Pgp) and multi-drug resistance-associated protein (MRP1), which act as drug efflux pumps.30,164, 165, 166, 167, 168 Furthermore, MVP is effective as an independent prognostic factor in predicting tumor response to standard chemotherapy and survival in patients with advanced stages of many cancers.169 However, no vault components have been identified to directly be involved or interact with the chemotherapeutic compounds.53
According to the surprising data obtained in a recent study, safe levels may be a good predictor of drug resistance, as their up-regulation alone is not sufficient to confer the drug-resistant phenotype. This indicates a need for extra factors to vault-mediated MDR. Vault up-regulation may be necessary but not sufficient for multidrug resistance.48 Previous studies showed the components that bind safely to cancer cells are resistant to chemotherapy drugs.30,48,170, 171, 172 Importantly, another previously discovered aspect of Persson et al. study found that svRNAb down-regulates cytochrome P450 3A4 (CYP3A4), a key enzyme in drug metabolism. These findings expand the repertoire of small regulatory RNAs and, for the first time, assign functions to vtRNAs that may help explain the relationship between vault particles and drug resistance.69 CYP3A4 is the most abundant cytochrome P450 enzyme in the human liver and intestine and contributes to the metabolism of >60% of all drugs.171,173 The hepatic expression levels of CYP3A4 vary widely between individuals.171 However, the detailed regulatory mechanisms of CYP3A4 expression remain unclear. Additionally, a negative correlation was found between svRNAb and CYP3A4 in human tissue samples.171
Furthermore, luciferase assays in human liver cancer G2 (HepG2) cells confirmed that svRNAb regulates CYP3A4 expression by directly targeting CYP3A4 and interacting with a validated binding site in the CYP3A4 3′UTR.171 The results provided insight into changes in CYP3A4 expression between persons and provided a new way to tailor individualized drug therapy. Moreover, this study provided a novel regulatory mechanism of svRNAb in MDR cells.171 New data suggest that vtRNA may be involved in sequestering chemotherapeutic compounds, preventing the drugs from reaching their target sites.56,62 Higher levels of vtRNA1 expression appear to be involved in the drug resistance phenotype than mitoxantrone.56,62 Interestingly, inhibition of vtRNA expression by RNA interference in resistant human cancer cells to chemotherapy resulted in a progressive decline in resistance.62
Consistent with the above results, vtRNA was found to be significantly superior to MVP by cloning the human gene for vtRNA and carefully determining the levels of MVP and vtRNA in MDR cells. Measurements of vault particle deposition in multiresistant cells showed a 15-fold increase in vault synthesis. The vaults synthesis directly related to multidrug resistance supports a direct role of vaults in drug resistancen.30 Besides, another group of researchers showed inhibition of nc886 increased chemosensitivity, induced apoptosis, and suppressed MVP protein expression, a key regulator of drug resistance in cervical cancer cells. Furthermore, they showed E2F transcription factor 1 (E2F1) promoted nc886 transcription and can promote drug resistance in cervical cancer cells by altering MVP expression.89,174
Ferro et al. described a new insight into the survival-promoting role of vtRNA1-1 in vitro and in vivo. Knockdown (KO) of vtRNA1-1 in human HCC cells inhibits transcription factor EB (TFEB) nuclear translocation leading to dysfunction of the lysosomal compartment, resulting in the down-regulation of coordinated lysosomal expression and regulation (CLEAR) network genes. In addition, the depletion of vtRNA1-1 increased the activation of the MAPK cascades responsible for inactivating TFEB, MAPK1/ERK2, and MAPK3/ERK1. Finally, the study group showed the absence of vtRNA1-1 reduced drug hemolysis and significantly inhibited tumor cell proliferation in vitro and in mouse models. These results demonstrate the role of vtRNA1-1 in maintaining the function of the intracellular catabolic compartment in human hepatocellular carcinoma cells and highlight its importance in lysosome-mediated chemotherapy resistance.137
Conclusion and perspectives
Most human RNA transcripts are not known to encode proteins, and ncRNAs are known to regulate cellular physiology and cellular functions. Rapid advances in network biology indicate a highly conserved evolutionary subset of ncRNAs are represented by vtRNAs, so named because they were first discovered as part of a larger ribonucleoprotein complex known as the vault. Although vtRNAs were first described 30 years ago, their molecular functions are still under investigation. In humans, the vtRNA-ncRNA family, consisting of only four members, plays a vital role in cells. As described in this review, vtRNAs are involved in cell proliferation, metabolic regulation of different classes of RNA, intracellular transport, nucleocytoplasmic transport, nuclear pore complex formation, regulation of central signaling pathways, and cellular communication. In addition, vtRNAs play a crucial role in immune responses, influence apoptosis, and autophagy, and are involved in detoxification processes, and mechanisms of resistance to many drugs. Most of our current knowledge of the functions of vtRNAs focuses on viral infections and cancer. What are the general biological principles of regulation of cellular processes by vtRNA? The relationship between vtRNA and cellular machinery requires further investigation, as the role of vtRNA is associated with vault particles, and the function of vault particles is not well understood. Interestingly, infection by several virus families, including γ-herpesviridae, paramyxoviruses, and influenza A, elicits a high expression of vtRNA. On the other hand, high expression of vault RNAs has been shown to lead to increased viral loads. Viruses are known to hijack cells and cellular replication machinery to maximize viral replication while inhibiting cellular defense mechanisms. We wonder if the vtRNA regulatory axis could be an important aspect of the molecular pathogenesis mechanism of the new lethal viral pandemic SARS-CoV-2. A recent study reported that SARS-CoV-2 infection induces autophagy and apoptosis in human microvascular bronchial endothelial and epithelial cells175 and can bypass antiviral pathways induced by viral double-stranded RNA, including PKR and interferon (IFN) signaling.176, 177, 178 So, could these strong observations point to a key role for vtRNAs in SARS-CoV-2 infection? Furthermore, the details on the identity, role, and mechanism of action of vtRNA will advance the mechanistic understanding of the molecular mechanisms of the RNA-directed in pathological conditions, as well as the development of new RNA-based diagnostic and therapeutic strategies in viral infections and unknown diseases. However, these results are still limited and future research in this area may lead to a better understanding of previously discovered processes.
Conflict of interests
The author declares that there is no conflict of interests.
Footnotes
Peer review under responsibility of Chongqing Medical University.
References
- 1.Hüttenhofer A., Schattner P., Polacek N. Non-coding RNAs: hope or hype? Trends Genet. 2005;21(5):289–297. doi: 10.1016/j.tig.2005.03.007. [DOI] [PubMed] [Google Scholar]
- 2.Landgraf P., Rusu M., Sheridan R., et al. A mammalian microRNA expression atlas based on small RNA library sequencing. Cell. 2007;129(7):1401–1414. doi: 10.1016/j.cell.2007.04.040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Mattick J.S. RNA regulation: a new genetics? Nat Rev Genet. 2004;5(4):316–323. doi: 10.1038/nrg1321. [DOI] [PubMed] [Google Scholar]
- 4.Wang Z., Gerstein M., Snyder M. RNA-Seq: a revolutionary tool for transcriptomics. Nat Rev Genet. 2009;10(1):57–63. doi: 10.1038/nrg2484. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Long Y., Wang X., Youmans D.T., et al. How do lncRNAs regulate transcription? Sci Adv. 2017;3(9) doi: 10.1126/sciadv.aao2110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Mattick J.S. Non-coding RNAs: the architects of eukaryotic complexity. EMBO Rep. 2001;2(11):986–991. doi: 10.1093/embo-reports/kve230. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Carninci P., Kasukawa T., Katayama S., et al. The transcriptional landscape of the mammalian genome. Science. 2005;309(5740):1559–1563. doi: 10.1126/science.1112014. [DOI] [PubMed] [Google Scholar]
- 8.Lander E.S., Linton L.M., Birren B., et al. Initial sequencing and analysis of the human genome. Nature. 2001;409(6822):860–921. doi: 10.1038/35057062. [DOI] [PubMed] [Google Scholar]
- 9.Venter J.C., Adams M.D., Myers E.W., et al. The sequence of the human genome. Science. 2001;291(5507):1304–1351. doi: 10.1126/science.1058040. [DOI] [PubMed] [Google Scholar]
- 10.Kickhoefer V.A., Stephen A.G., Harrington L., et al. Vaults and telomerase share a common subunit, TEP1. J Biol Chem. 1999;274(46):32712–32717. doi: 10.1074/jbc.274.46.32712. [DOI] [PubMed] [Google Scholar]
- 11.Min H., Yoon S. Got target? Computational methods for microRNA target prediction and their extension. Exp Mol Med. 2010;42(4):233–244. doi: 10.3858/emm.2010.42.4.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Mattick J.S., Makunin I.V. Non-coding RNA. Hum Mol Genet. 2006;15 Spec(1):R17–R29. doi: 10.1093/hmg/ddl046. [DOI] [PubMed] [Google Scholar]
- 13.Eddy S.R. Non-coding RNA genes and the modern RNA world. Nat Rev Genet. 2001;2(12):919–929. doi: 10.1038/35103511. [DOI] [PubMed] [Google Scholar]
- 14.Tiedge H., Chen W., Brosius J. Primary structure, neural-specific expression, and dendritic location of human BC200 RNA. J Neurosci. 1993;13(6):2382–2390. doi: 10.1523/JNEUROSCI.13-06-02382.1993. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Lerner M.R., Boyle J.A., Hardin J.A., et al. Two novel classes of small ribonucleoproteins detected by antibodies associated with lupus erythematosus. Science. 1981;211(4480):400–402. doi: 10.1126/science.6164096. [DOI] [PubMed] [Google Scholar]
- 16.Scott W.G. Ribozymes. Curr Opin Struct Biol. 2007;17(3):280–286. doi: 10.1016/j.sbi.2007.05.003. [DOI] [PubMed] [Google Scholar]
- 17.Amaral P.P., Dinger M.E., Mercer T.R., et al. The eukaryotic genome as an RNA machine. Science. 2008;319(5871):1787–1789. doi: 10.1126/science.1155472. [DOI] [PubMed] [Google Scholar]
- 18.Gebetsberger J., Polacek N. Slicing tRNAs to boost functional ncRNA diversity. RNA Biol. 2013;10(12):1798–1806. doi: 10.4161/rna.27177. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Sabin L.R., Delás M.J., Hannon G.J. Dogma derailed: the many influences of RNA on the genome. Mol Cell. 2013;49(5):783–794. doi: 10.1016/j.molcel.2013.02.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Tuck A.C., Tollervey D. RNA in pieces. Trends Genet. 2011;27(10):422–432. doi: 10.1016/j.tig.2011.06.001. [DOI] [PubMed] [Google Scholar]
- 21.Hahne J.C., Lampis A., Valeri N. Vault RNAs: hidden gems in RNA and protein regulation. Cell Mol Life Sci. 2021;78(4):1487–1499. doi: 10.1007/s00018-020-03675-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Kedersha N.L., Rome L.H. Isolation and characterization of a novel ribonucleoprotein particle: large structures contain a single species of small RNA. J Cell Biol. 1986;103(3):699–709. doi: 10.1083/jcb.103.3.699. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Kickhoefer V.A., Searles R.P., Kedersha N.L., et al. Vault ribonucleoprotein particles from rat and bullfrog contain a related small RNA that is transcribed by RNA polymerase III. J Biol Chem. 1993;268(11):7868–7873. [PubMed] [Google Scholar]
- 24.Vilalta A., Kickhoefer V.A., Rome L.H., et al. The rat vault RNA gene contains a unique RNA polymerase III promoter composed of both external and internal elements that function synergistically. J Biol Chem. 1994;269(47):29752–29759. [PubMed] [Google Scholar]
- 25.Chugani D.C., Rome L.H., Kedersha N.L. Evidence that vault ribonucleoprotein particles localize to the nuclear pore complex. J Cell Sci. 1993;106(Pt 1):23–29. doi: 10.1242/jcs.106.1.23. [DOI] [PubMed] [Google Scholar]
- 26.Suprenant K.A. Vault ribonucleoprotein particles: sarcophagi, gondolas, or safety deposit boxes? Biochemistry. 2002;41(49):14447–14454. doi: 10.1021/bi026747e. [DOI] [PubMed] [Google Scholar]
- 27.Kedersha N.L., Heuser J.E., Chugani D.C., et al. Vaults. III. Vault ribonucleoprotein particles open into flower-like structures with octagonal symmetry. J Cell Biol. 1991;112(2):225–235. doi: 10.1083/jcb.112.2.225. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Stephen A.G., Raval-Fernandes S., Huynh T., et al. Assembly of vault-like particles in insect cells expressing only the major vault protein. J Biol Chem. 2001;276(26):23217–23220. doi: 10.1074/jbc.C100226200. [DOI] [PubMed] [Google Scholar]
- 29.van Zon A., Mossink M.H., Scheper R.J., et al. The vault complex. Cell Mol Life Sci. 2003;60(9):1828–1837. doi: 10.1007/s00018-003-3030-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Kickhoefer V.A., Rajavel K.S., Scheffer G.L., et al. Vaults are up-regulated in multidrug-resistant cancer cell lines. J Biol Chem. 1998;273(15):8971–8974. doi: 10.1074/jbc.273.15.8971. [DOI] [PubMed] [Google Scholar]
- 31.Abbondanza C., Rossi V., Roscigno A., et al. Interaction of vault particles with estrogen receptor in the MCF-7 breast cancer cell. J Cell Biol. 1998;141(6):1301–1310. doi: 10.1083/jcb.141.6.1301. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.van Zon A., Mossink M.H., Schoester M., et al. Multiple human vault RNAs. Expression and association with the vault complex. J Biol Chem. 2001;276(40):37715–37721. doi: 10.1074/jbc.M106055200. [DOI] [PubMed] [Google Scholar]
- 33.Hamill D.R., Suprenant K.A. Characterization of the sea urchin major vault protein: a possible role for vault ribonucleoprotein particles in nucleocytoplasmic transport. Dev Biol. 1997;190(1):117–128. doi: 10.1006/dbio.1997.8676. [DOI] [PubMed] [Google Scholar]
- 34.Herrmann C., Golkaramnay E., Inman E., et al. Recombinant major vault protein is targeted to neuritic tips of PC12 cells. J Cell Biol. 1999;144(6):1163–1172. doi: 10.1083/jcb.144.6.1163. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Herrmann C., Volknandt W., Wittich B., et al. The major vault protein (MVP100) is contained in cholinergic nerve terminals of electric ray electric organ. J Biol Chem. 1996;271(23):13908–13915. doi: 10.1074/jbc.271.23.13908. [DOI] [PubMed] [Google Scholar]
- 36.Li J.Y., Volknandt W., Dahlstrom A., et al. Axonal transport of ribonucleoprotein particles (vaults) Neuroscience. 1999;91(3):1055–1065. doi: 10.1016/s0306-4522(98)00622-8. [DOI] [PubMed] [Google Scholar]
- 37.Nandy C., Mrázek J., Stoiber H., et al. Epstein-Barr virus-induced expression of a novel human vault RNA. J Mol Biol. 2009;388(4):776–784. doi: 10.1016/j.jmb.2009.03.031. [DOI] [PubMed] [Google Scholar]
- 38.Kong L.B., Siva A.C., Kickhoefer V.A., et al. RNA location and modeling of a WD40 repeat domain within the vault. RNA. 2000;6(6):890–900. doi: 10.1017/s1355838200000157. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Berger W., Steiner E., Grusch M., et al. Vaults and the major vault protein: novel roles in signal pathway regulation and immunity. Cell Mol Life Sci. 2009;66(1):43–61. doi: 10.1007/s00018-008-8364-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Kickhoefer V.A., Liu Y., Kong L.B., et al. The Telomerase/vault-associated protein TEP1 is required for vault RNA stability and its association with the vault particle. J Cell Biol. 2001;152(1):157–164. doi: 10.1083/jcb.152.1.157. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Stadler P.F., Chen J.J.L., Hackermüller J., et al. Evolution of vault RNAs. Mol Biol Evol. 2009;26(9):1975–1991. doi: 10.1093/molbev/msp112. [DOI] [PubMed] [Google Scholar]
- 42.Gallo S., Kong E., Ferro I., et al. Small but powerful: the human vault RNAs as multifaceted modulators of pro-survival characteristics and tumorigenesis. Cancers. 2022;14(11):2787. doi: 10.3390/cancers14112787. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Mrázek J., Kreutmayer S.B., Grässer F.A., et al. Subtractive hybridization identifies novel differentially expressed ncRNA species in EBV-infected human B cells. Nucleic Acids Res. 2007;35(10):e73. doi: 10.1093/nar/gkm244. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Fort R.S., Garat B., Sotelo-Silveira J.R., et al. vtRNA2-1/nc886 produces a small RNA that contributes to its tumor suppression action through the microRNA pathway in prostate cancer. Noncoding RNA. 2020;6(1):7. doi: 10.3390/ncrna6010007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Griffiths-Jones S., Grocock R.J., van Dongen S., et al. miRBase: microRNA sequences, targets and gene nomenclature. Nucleic Acids Res. 2006;34(Database issue):D140–D144. doi: 10.1093/nar/gkj112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Willis I.M. RNA polymerase III. Genes, factors and transcriptional specificity. Eur J Biochem. 1993;212(1):1–11. doi: 10.1111/j.1432-1033.1993.tb17626.x. [DOI] [PubMed] [Google Scholar]
- 47.Márquez Contreras E., Casado Martínez J.J., Corchado Albalat Y., et al. Efficacy of an intervention to improve treatment compliance in hyperlipidemias. Atención Primaria. 2004;33(8):443–450. doi: 10.1016/S0212-6567(04)79430-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Siva A.C., Raval-Fernandes S., Stephen A.G., et al. Up-regulation of vaults may be necessary but not sufficient for multidrug resistance. Int J Cancer. 2001;92(2):195–202. doi: 10.1002/1097-0215(200102)9999:9999<::aid-ijc1168>3.0.co;2-7. [DOI] [PubMed] [Google Scholar]
- 49.Stewart P.L., Makabi M., Lang J., et al. Sea urchin vault structure, composition, and differential localization during development. BMC Dev Biol. 2005;5:3. doi: 10.1186/1471-213X-5-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Kolev N.G., Rajan K.S., Tycowski K.T., et al. The vault RNA of Trypanosoma brucei plays a role in the production of trans-spliced mRNA. J Biol Chem. 2019;294(43):15559–15574. doi: 10.1074/jbc.RA119.008580. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Sudha G., Yamunadevi S., Tyagi N., et al. Structural and molecular basis of interaction of HCV non-structural protein 5A with human casein kinase 1α and PKR. BMC Struct Biol. 2012;12:28. doi: 10.1186/1472-6807-12-28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Li F., Chen Y., Zhang Z., et al. Robust expression of vault RNAs induced by influenza A virus plays a critical role in suppression of PKR-mediated innate immunity. Nucleic Acids Res. 2015;43(21):10321–10337. doi: 10.1093/nar/gkv1078. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Gopinath S.C.B., Matsugami A., Katahira M., et al. Human vault-associated non-coding RNAs bind to mitoxantrone, a chemotherapeutic compound. Nucleic Acids Res. 2005;33(15):4874–4881. doi: 10.1093/nar/gki809. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Kickhoefer V.A., Poderycki M.J., Chan E.K.L., et al. The La RNA-binding protein interacts with the vault RNA and is a vault-associated protein. J Biol Chem. 2002;277(43):41282–41286. doi: 10.1074/jbc.M206980200. [DOI] [PubMed] [Google Scholar]
- 55.Lee K.S., Park J.L., Lee K., et al. nc886, a non-coding RNA of anti-proliferative role, is suppressed by CpG DNA methylation in human gastric cancer. Oncotarget. 2014;5(11):3944–3955. doi: 10.18632/oncotarget.2047. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Chen J., OuYang H., An X., et al. Vault RNAs partially induces drug resistance of human tumor cells MCF-7 by binding to the RNA/DNA-binding protein PSF and inducing oncogene GAGE6. PLoS One. 2018;13(1) doi: 10.1371/journal.pone.0191325. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 57.Scheffer G.L., Schroeijers A.B., Izquierdo M.A., et al. Lung resistance-related protein/major vault protein and vaults in multidrug-resistant cancer. Curr Opin Oncol. 2000;12(6):550–556. doi: 10.1097/00001622-200011000-00007. [DOI] [PubMed] [Google Scholar]
- 58.Scheper R.J., Broxterman H.J., Scheffer G.L., et al. Overexpression of a M(r) 110, 000 vesicular protein in non-P-glycoprotein-mediated multidrug resistance. Cancer Res. 1993;53(7):1475–1479. [PubMed] [Google Scholar]
- 59.Izquierdo M.A., Scheffer G.L., Flens M.J., et al. Broad distribution of the multidrug resistance-related vault lung resistance protein in normal human tissues and tumors. Am J Pathol. 1996;148(3):877–887. [PMC free article] [PubMed] [Google Scholar]
- 60.Izquierdo M.A., Shoemaker R.H., Flens M.J., et al. Overlapping phenotypes of multidrug resistance among panels of human cancer-cell lines. Int J Cancer. 1996;65(2):230–237. doi: 10.1002/(SICI)1097-0215(19960117)65:2<230::AID-IJC17>3.0.CO;2-H. [DOI] [PubMed] [Google Scholar]
- 61.Kitazono M., Sumizawa T., Takebayashi Y., et al. Multidrug resistance and the lung resistance-related protein in human colon carcinoma SW-620 cells. J Natl Cancer Inst. 1999;91(19):1647–1653. doi: 10.1093/jnci/91.19.1647. [DOI] [PubMed] [Google Scholar]
- 62.Gopinath S.C.B., Wadhwa R., Kumar P.K.R. Expression of noncoding vault RNA in human malignant cells and its importance in mitoxantrone resistance. Mol Cancer Res. 2010;8(11):1536–1546. doi: 10.1158/1541-7786.MCR-10-0242. [DOI] [PubMed] [Google Scholar]
- 63.Chung J.H., Ginn-Pease M.E., Eng C. Phosphatase and tensin homologue deleted on chromosome 10 (PTEN) has nuclear localization signal-like sequences for nuclear import mediated by major vault protein. Cancer Res. 2005;65(10):4108–4116. doi: 10.1158/0008-5472.CAN-05-0124. [DOI] [PubMed] [Google Scholar]
- 64.Kim E., Lee S., Mian M.F., et al. Crosstalk between Src and major vault protein in epidermal growth factor-dependent cell signalling. FEBS J. 2006;273(4):793–804. doi: 10.1111/j.1742-4658.2006.05112.x. [DOI] [PubMed] [Google Scholar]
- 65.Ryu S.J., An H.J., Oh Y.S., et al. On the role of major vault protein in the resistance of senescent human diploid fibroblasts to apoptosis. Cell Death Differ. 2008;15(11):1673–1680. doi: 10.1038/cdd.2008.96. [DOI] [PubMed] [Google Scholar]
- 66.Amort M., Nachbauer B., Tuzlak S., et al. Expression of the vault RNA protects cells from undergoing apoptosis. Nat Commun. 2015;6:7030. doi: 10.1038/ncomms8030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Kowalski M.P., Dubouix-Bourandy A., Bajmoczi M., et al. Host resistance to lung infection mediated by major vault protein in epithelial cells. Science. 2007;317(5834):130–132. doi: 10.1126/science.1142311. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Horos R., Büscher M., Kleinendorst R., et al. The small non-coding vault RNA1-1 acts as a riboregulator of autophagy. Cell. 2019;176(5):1054–1067. doi: 10.1016/j.cell.2019.01.030. e12. [DOI] [PubMed] [Google Scholar]
- 69.Persson H., Kvist A., Vallon-Christersson J., et al. The non-coding RNA of the multidrug resistance-linked vault particle encodes multiple regulatory small RNAs. Nat Cell Biol. 2009;11(10):1268–1271. doi: 10.1038/ncb1972. [DOI] [PubMed] [Google Scholar]
- 70.Bracher L., Ferro I., Pulido-Quetglas C., et al. Human vtRNA1-1 levels modulate signaling pathways and regulate apoptosis in human cancer cells. Biomolecules. 2020;10(4):614. doi: 10.3390/biom10040614. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Fontenla S., Langleib M., de la Torre-Escudero E., et al. Role of Fasciola hepatica small RNAs in the interaction with the mammalian host. Front Cell Infect Microbiol. 2021;11:812141. doi: 10.3389/fcimb.2021.812141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Umu S.U., Langseth H., Bucher-Johannessen C., et al. A comprehensive profile of circulating RNAs in human serum. RNA Biol. 2018;15(2):242–250. doi: 10.1080/15476286.2017.1403003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Szilágyi M., Pös O., Márton É., et al. Circulating cell-free nucleic acids: main characteristics and clinical application. Int J Mol Sci. 2020;21(18):6827. doi: 10.3390/ijms21186827. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Lefebvre F.A., Benoit Bouvrette L.P., Perras L., et al. Comparative transcriptomic analysis of human and Drosophila extracellular vesicles. Sci Rep. 2016;6:27680. doi: 10.1038/srep27680. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Mesquita-Ribeiro R., Fort R.S., Rathbone A., et al. Distinct small non-coding RNA landscape in the axons and released extracellular vesicles of developing primary cortical neurons and the axoplasm of adult nerves. RNA Biol. 2021;18(sup2):832–855. doi: 10.1080/15476286.2021.2000792. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Nolte-'t Hoen E.N.M., Buermans H.P.J., Waasdorp M., et al. Deep sequencing of RNA from immune cell-derived vesicles uncovers the selective incorporation of small non-coding RNA biotypes with potential regulatory functions. Nucleic Acids Res. 2012;40(18):9272–9285. doi: 10.1093/nar/gks658. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Romanelli V., Nakabayashi K., Vizoso M., et al. Variable maternal methylation overlapping the nc886/vtRNA2-1 locus is locked between hypermethylated repeats and is frequently altered in cancer. Epigenetics. 2014;9(5):783–790. doi: 10.4161/epi.28323. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.You J.S., Jones P.A. Cancer genetics and epigenetics: two sides of the same coin? Cancer Cell. 2012;22(1):9–20. doi: 10.1016/j.ccr.2012.06.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Lu Y., Chan Y.T., Tan H.Y., et al. Epigenetic regulation in human cancer: the potential role of epi-drug in cancer therapy. Mol Cancer. 2020;19:79. doi: 10.1186/s12943-020-01197-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Kumar S., Gonzalez E.A., Rameshwar P., et al. Non-coding RNAs as mediators of epigenetic changes in malignancies. Cancers. 2020;12(12):3657. doi: 10.3390/cancers12123657. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Bijnsdorp I.V., van Royen M.E., Verhaegh G.W., et al. The non-coding transcriptome of prostate cancer: implications for clinical practice. Mol Diagn Ther. 2017;21(4):385–400. doi: 10.1007/s40291-017-0271-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Bolton E.M., Tuzova A.V., Walsh A.L., et al. Noncoding RNAs in prostate cancer: the long and the short of it. Clin Cancer Res. 2014;20(1):35–43. doi: 10.1158/1078-0432.CCR-13-1989. [DOI] [PubMed] [Google Scholar]
- 83.Nana-Sinkam S.P., Croce C.M. Non-coding RNAs in cancer initiation and progression and as novel biomarkers. Mol Oncol. 2011;5(6):483–491. doi: 10.1016/j.molonc.2011.10.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Yan H., Bu P. Non-coding RNA in cancer. Essays Biochem. 2021;65(4):625–639. doi: 10.1042/EBC20200032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Mouraviev V., Lee B., Patel V., et al. Clinical prospects of long noncoding RNAs as novel biomarkers and therapeutic targets in prostate cancer. Prostate Cancer Prostatic Dis. 2016;19(1):14–20. doi: 10.1038/pcan.2015.48. [DOI] [PubMed] [Google Scholar]
- 86.Treppendahl M.B., Qiu X., Søgaard A., et al. Allelic methylation levels of the noncoding VTRNA2-1 located on chromosome 5q31.1 predict outcome in AML. Blood. 2012;119(1):206–216. doi: 10.1182/blood-2011-06-362541. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Helbo A.S., Treppendahl M., Aslan D., et al. Hypermethylation of the VTRNA1-3 promoter is associated with poor outcome in lower risk myelodysplastic syndrome patients. Genes. 2015;6(4):977–990. doi: 10.3390/genes6040977. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Kunkeaw N., Jeon S.H., Lee K., et al. Cell death/proliferation roles for nc886, a non-coding RNA, in the protein kinase R pathway in cholangiocarcinoma. Oncogene. 2013;32(32):3722–3731. doi: 10.1038/onc.2012.382. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Lee K., Kunkeaw N., Jeon S.H., et al. Precursor miR-886, a novel noncoding RNA repressed in cancer, associates with PKR and modulates its activity. RNA. 2011;17(6):1076–1089. doi: 10.1261/rna.2701111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Yu M.C., Lee C.W., Lin C.H., et al. Differential hypermethylation of the VTRNA2-1 promoter in hepatocellular carcinoma as a prognostic factor: tumor marker prognostic study. Int J Surg. 2020;79:282–289. doi: 10.1016/j.ijsu.2020.05.016. [DOI] [PubMed] [Google Scholar]
- 91.Henderson A.R., Wang Q., Meechoovet B., et al. DNA methylation and expression profiles of whole blood in Parkinson's disease. Front Genet. 2021;12:640266. doi: 10.3389/fgene.2021.640266. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Kumaresan M., Khan S. Spectrum of non-motor symptoms in Parkinson's disease. Cureus. 2021;13(2) doi: 10.7759/cureus.13275. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Wang C., Chen L., Yang Y., et al. Identification of potential blood biomarkers for Parkinson's disease by gene expression and DNA methylation data integration analysis. Clin Epigenet. 2019;11:24. doi: 10.1186/s13148-019-0621-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Miñones-Moyano E., Friedländer M.R., Pallares J., et al. Upregulation of a small vault RNA (svtRNA2-1a) is an early event in Parkinson disease and induces neuronal dysfunction. RNA Biol. 2013;10(7):1093–1106. doi: 10.4161/rna.24813. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Wakatsuki S., Araki T. Novel molecular basis for synapse formation: small non-coding vault RNA functions as a riboregulator of MEK1 to modulate synaptogenesis. Front Mol Neurosci. 2021;14:748721. doi: 10.3389/fnmol.2021.748721. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Wakatsuki S., Ohno M., Araki T. Human vault RNA1-1, but not vault RNA2-1, modulates synaptogenesis. Commun Integr Biol. 2021;14(1):61–65. doi: 10.1080/19420889.2021.1909229. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Manandhar B., Ahn J.M. Glucagon-like peptide-1 (GLP-1) analogs: recent advances, new possibilities, and therapeutic implications. J Med Chem. 2015;58(3):1020–1037. doi: 10.1021/jm500810s. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Lin C.H., Lee Y.S., Huang Y.Y., et al. Methylation status of vault RNA 2-1 promoter is a predictor of glycemic response to glucagon-like peptide-1 analog therapy in type 2 diabetes mellitus. BMJ Open Diabetes Res Care. 2021;9(1) doi: 10.1136/bmjdrc-2020-001416. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Sallustio F., Curci C., di Leo V., et al. A new vision of IgA nephropathy: the missing link. Int J Mol Sci. 2019;21(1):189. doi: 10.3390/ijms21010189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Sallustio F., Serino G., Cox S.N., et al. Aberrantly methylated DNA regions lead to low activation of CD4+ T-cells in IgA nephropathy. Clin Sci. 2016;130(9):733–746. doi: 10.1042/CS20150711. [DOI] [PubMed] [Google Scholar]
- 101.Li M., Yu X. Genetic study of immunoglobulin A nephropathy: from research to clinical application. Nephrology. 2018;23(Suppl 4):26–31. doi: 10.1111/nep.13470. [DOI] [PubMed] [Google Scholar]
- 102.D'Orazio J., Jarrett S., Amaro-Ortiz A., et al. UV radiation and the skin. Int J Mol Sci. 2013;14(6):12222–12248. doi: 10.3390/ijms140612222. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Gromkowska-Kępka K.J., Puścion-Jakubik A., Markiewicz-Żukowska R., et al. The impact of ultraviolet radiation on skin photoaging - review of in vitro studies. J Cosmet Dermatol. 2021;20(11):3427–3431. doi: 10.1111/jocd.14033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Panich U., Sittithumcharee G., Rathviboon N., et al. Ultraviolet radiation-induced skin aging: the role of DNA damage and oxidative stress in epidermal stem cell damage mediated skin aging. Stem Cell Int. 2016;2016:7370642. doi: 10.1155/2016/7370642. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Lee K.S., Shin S., Cho E., et al. nc886, a non-coding RNA, inhibits UVB-induced MMP-9 and COX-2 expression via the PKR pathway in human keratinocytes. Biochem Biophys Res Commun. 2019;512(4):647–652. doi: 10.1016/j.bbrc.2019.01.068. [DOI] [PubMed] [Google Scholar]
- 106.Maginnis M.S. Virus-receptor interactions: the key to cellular invasion. J Mol Biol. 2018;430(17):2590–2611. doi: 10.1016/j.jmb.2018.06.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Bagga S., Bouchard M.J. Cell cycle regulation during viral infection. Methods Mol Biol. 2014;1170:165–227. doi: 10.1007/978-1-4939-0888-2_10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Liu W., Ding C. Roles of LncRNAs in viral infections. Front Cell Infect Microbiol. 2017;7:205. doi: 10.3389/fcimb.2017.00205. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Fort R.S., Duhagon M.A. Pan-cancer chromatin analysis of the human vtRNA genes uncovers their association with cancer biology. F1000Res. 2021;10:182. doi: 10.12688/f1000research.28510.1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Motsch N., Pfuhl T., Mrazek J., et al. Epstein-Barr virus-encoded latent membrane protein 1 (LMP1) induces the expression of the cellular microRNA miR-146a. RNA Biol. 2007;4(3):131–137. doi: 10.4161/rna.4.3.5206. [DOI] [PubMed] [Google Scholar]
- 111.Weitzman M.D., Fradet-Turcotte A. Virus DNA replication and the host DNA damage response. Annu Rev Virol. 2018;5(1):141–164. doi: 10.1146/annurev-virology-092917-043534. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Meurs E., Chong K., Galabru J., et al. Molecular cloning and characterization of the human double-stranded RNA-activated protein kinase induced by interferon. Cell. 1990;62(2):379–390. doi: 10.1016/0092-8674(90)90374-n. [DOI] [PubMed] [Google Scholar]
- 113.de Haro C., Méndez R., Santoyo J. The eIF-2 alpha kinases and the control of protein synthesis. Faseb J. 1996;10(12):1378–1387. doi: 10.1096/fasebj.10.12.8903508. [DOI] [PubMed] [Google Scholar]
- 114.Kumar A., Haque J., Lacoste J., et al. Double-stranded RNA-dependent protein kinase activates transcription factor NF-kappa B by phosphorylating I kappa B. Proc Natl Acad Sci U S A. 1994;91(14):6288–6292. doi: 10.1073/pnas.91.14.6288. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Sharma S., TenOever B.R., Grandvaux N., et al. Triggering the interferon antiviral response through an IKK-related pathway. Science. 2003;300(5622):1148–1151. doi: 10.1126/science.1081315. [DOI] [PubMed] [Google Scholar]
- 116.Schneider W.M., Chevillotte M.D., Rice C.M. Interferon-stimulated genes: a complex web of host defenses. Annu Rev Immunol. 2014;32:513–545. doi: 10.1146/annurev-immunol-032713-120231. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Katze M.G., Tomita J., Black T., et al. Influenza virus regulates protein synthesis during infection by repressing autophosphorylation and activity of the cellular 68, 000-Mr protein kinase. J Virol. 1988;62(10):3710–3717. doi: 10.1128/jvi.62.10.3710-3717.1988. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Lu Y., Wambach M., Katze M.G., et al. Binding of the influenza virus NS1 protein to double-stranded RNA inhibits the activation of the protein kinase that phosphorylates the elF-2 translation initiation factor. Virology. 1995;214(1):222–228. doi: 10.1006/viro.1995.9937. [DOI] [PubMed] [Google Scholar]
- 119.Lee T.G., Tang N., Thompson S., et al. The 58, 000-dalton cellular inhibitor of the interferon-induced double-stranded RNA-activated protein kinase (PKR) is a member of the tetratricopeptide repeat family of proteins. Mol Cell Biol. 1994;14(4):2331–2342. doi: 10.1128/mcb.14.4.2331. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Tan S.L., Katze M.G. Biochemical and genetic evidence for complex formation between the influenza A virus NS1 protein and the interferon-induced PKR protein kinase. J Interferon Cytokine Res. 1998;18(9):757–766. doi: 10.1089/jir.1998.18.757. [DOI] [PubMed] [Google Scholar]
- 121.Melville M.W., Tan S.L., Wambach M., et al. The cellular inhibitor of the PKR protein kinase, P58IPK, is an influenza virus-activated co-chaperone that modulates heat shock protein 70 activity. J Biol Chem. 1999;274(6):3797–3803. doi: 10.1074/jbc.274.6.3797. [DOI] [PubMed] [Google Scholar]
- 122.Tan S.L., Gale M.J., Jr., Katze M.G. Double-stranded RNA-independent dimerization of interferon-induced protein kinase PKR and inhibition of dimerization by the cellular P58IPK inhibitor. Mol Cell Biol. 1998;18(5):2431–2443. doi: 10.1128/mcb.18.5.2431. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Bergmann M., Garcia-Sastre A., Carnero E., et al. Influenza virus NS1 protein counteracts PKR-mediated inhibition of replication. J Virol. 2000;74(13):6203–6206. doi: 10.1128/jvi.74.13.6203-6206.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Chien C.Y., Xu Y., Xiao R., et al. Biophysical characterization of the complex between double-stranded RNA and the N-terminal domain of the NS1 protein from influenza A virus: evidence for a novel RNA-binding mode. Biochemistry. 2004;43(7):1950–1962. doi: 10.1021/bi030176o. [DOI] [PubMed] [Google Scholar]
- 125.Min J.Y., Krug R.M. The primary function of RNA binding by the influenza A virus NS1 protein in infected cells: inhibiting the 2'-5' oligo (A) synthetase/RNase L pathway. Proc Natl Acad Sci U S A. 2006;103(18):7100–7105. doi: 10.1073/pnas.0602184103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Falcón A.M., Fortes P., Marión R.M., et al. Interaction of influenza virus NS1 protein and the human homologue of Staufen in vivo and in vitro. Nucleic Acids Res. 1999;27(11):2241–2247. doi: 10.1093/nar/27.11.2241. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Khaperskyy D.A., Hatchette T.F., McCormick C. Influenza A virus inhibits cytoplasmic stress granule formation. Faseb J. 2012;26(4):1629–1639. doi: 10.1096/fj.11-196915. [DOI] [PubMed] [Google Scholar]
- 128.Jeon S.H., Lee K., Lee K.S., et al. Characterization of the direct physical interaction of nc886, a cellular non-coding RNA, and PKR. FEBS Lett. 2012;586(19):3477–3484. doi: 10.1016/j.febslet.2012.07.076. [DOI] [PubMed] [Google Scholar]
- 129.Klionsky D.J., Abdelmohsen K., Abe A., et al. Guidelines for the use and interpretation of assays for monitoring autophagy (3rd edition) Autophagy. 2016;12(1):1–222. doi: 10.1080/15548627.2015.1100356. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Kroemer G., Mariño G., Levine B. Autophagy and the integrated stress response. Mol Cell. 2010;40(2):280–293. doi: 10.1016/j.molcel.2010.09.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Büscher M., Horos R., Haubrich K., et al. Mechanism of riboregulation of p62 protein oligomerisation by vault RNA1-1 in selective autophagy. bioRxiv. 2021;12:2021–2104. [Google Scholar]
- 132.Bjørkøy G., Lamark T., Brech A., et al. p62/SQSTM1 forms protein aggregates degraded by autophagy and has a protective effect on huntingtin-induced cell death. J Cell Biol. 2005;171(4):603–614. doi: 10.1083/jcb.200507002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Lim J., Lachenmayer M.L., Wu S., et al. Proteotoxic stress induces phosphorylation of p62/SQSTM1 by ULK1 to regulate selective autophagic clearance of protein aggregates. PLoS Genet. 2015;11(2) doi: 10.1371/journal.pgen.1004987. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Jakobi A.J., Huber S.T., Mortensen S.A., et al. Structural basis of p62/SQSTM1 helical filaments and their role in cellular cargo uptake. Nat Commun. 2020;11:440. doi: 10.1038/s41467-020-14343-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Oh J.E., Lee H.K. Autophagy as an innate immune modulator. Immune Netw. 2013;13(1):1–9. doi: 10.4110/in.2013.13.1.1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Büscher M., Horos R., Hentze M.W. ‘High vault-age': non-coding RNA control of autophagy. Open Biol. 2020;10(2):190307. doi: 10.1098/rsob.190307. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Ferro I., Gavini J., Gallo S., et al. The human vault RNA enhances tumorigenesis and chemoresistance through the lysosome in hepatocellular carcinoma. Autophagy. 2022;18(1):191–203. doi: 10.1080/15548627.2021.1922983. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Johansen T. Selective autophagy: RNA comes from the vault to regulate p62/SQSTM1. Curr Biol. 2019;29(8):R297–R299. doi: 10.1016/j.cub.2019.03.008. [DOI] [PubMed] [Google Scholar]
- 139.Horos R., Büscher M., Sachse C., et al. Vault RNA emerges as a regulator of selective autophagy. Autophagy. 2019;15(8):1463–1464. doi: 10.1080/15548627.2019.1609861. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Lee S.J., Pfluger P.T., Kim J.Y., et al. A functional role for the p62-ERK1 axis in the control of energy homeostasis and adipogenesis. EMBO Rep. 2010;11(3):226–232. doi: 10.1038/embor.2010.7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Duran A., Amanchy R., Linares J.F., et al. p62 is a key regulator of nutrient sensing in the mTORC1 pathway. Mol Cell. 2011;44:134–146. doi: 10.1016/j.molcel.2011.06.038. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Katsuragi Y., Ichimura Y., Komatsu M. p62/SQSTM1 functions as a signaling hub and an autophagy adaptor. FEBS J. 2015;282(24):4672–4678. doi: 10.1111/febs.13540. [DOI] [PubMed] [Google Scholar]
- 143.Moscat J., Karin M., Diaz-Meco M.T. p62 in cancer: signaling adaptor beyond autophagy. Cell. 2016;167(3):606–609. doi: 10.1016/j.cell.2016.09.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Büscher M., Horos R., Huppertz I., et al. Vault RNA1-1 riboregulates the autophagic function of p62 by binding to lysine 7 and arginine 21, both of which are critical for p62 oligomerization. RNA. 2022;28(5):742–755. doi: 10.1261/rna.079129.122. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Lamark T., Perander M., Outzen H., et al. Interaction codes within the family of mammalian phox and Bem1p domain-containing proteins. J Biol Chem. 2003;278(36):34568–34581. doi: 10.1074/jbc.M303221200. [DOI] [PubMed] [Google Scholar]
- 146.Obeng E. Apoptosis (programmed cell death) and its signals - a review. Braz J Biol. 2021;81(4):1133–1143. doi: 10.1590/1519-6984.228437. [DOI] [PubMed] [Google Scholar]
- 147.Elmore S. Apoptosis: a review of programmed cell death. Toxicol Pathol. 2007;35(4):495–516. doi: 10.1080/01926230701320337. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Zhang L., Li C., Su X. Emerging impact of the long noncoding RNA MIR22HG on proliferation and apoptosis in multiple human cancers. J Exp Clin Cancer Res. 2020;39:271. doi: 10.1186/s13046-020-01784-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Lv K., Liu Y., Zheng Y., et al. Long non-coding RNA MALAT1 regulates cell proliferation and apoptosis via miR-135b-5p/GPNMB axis in Parkinson's disease cell model. Biol Res. 2021;54:10. doi: 10.1186/s40659-021-00332-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Kong L., Hao Q., Wang Y., et al. Regulation of p53 expression and apoptosis by vault RNA2-1-5p in cervical cancer cells. Oncotarget. 2015;6(29):28371–28388. doi: 10.18632/oncotarget.4948. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Lei J., Xiao J.H., Zhang S.H., et al. Non-coding RNA 886 promotes renal cell carcinoma growth and metastasis through the Janus kinase 2/signal transducer and activator of transcription 3 signaling pathway. Mol Med Rep. 2017;16(4):4273–4278. doi: 10.3892/mmr.2017.7093. [DOI] [PubMed] [Google Scholar]
- 152.McElhinney J.M.W.R., Hasan A., Sajini A.A. The epitranscriptome landscape of small noncoding RNAs in stem cells. Stem Cell. 2020;38(10):1216–1228. doi: 10.1002/stem.3233. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.de Lucas B., Pérez L.M., Gálvez B.G. Importance and regulation of adult stem cell migration. J Cell Mol Med. 2018;22(2):746–754. doi: 10.1111/jcmm.13422. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Huang G., Ye S., Zhou X., et al. Molecular basis of embryonic stem cell self-renewal: from signaling pathways to pluripotency network. Cell Mol Life Sci. 2015;72(9):1741–1757. doi: 10.1007/s00018-015-1833-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Blanco S., Kurowski A., Nichols J., et al. The RNA-methyltransferase Misu (NSun2) poises epidermal stem cells to differentiate. PLoS Genet. 2011;7(12) doi: 10.1371/journal.pgen.1002403. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Morasso M.I., Tomic-Canic M. Epidermal stem cells: the cradle of epidermal determination, differentiation and wound healing. Biol Cell. 2005;97(3):173–183. doi: 10.1042/BC20040098. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Blanco S., Bandiera R., Popis M., et al. Stem cell function and stress response are controlled by protein synthesis. Nature. 2016;534(7607):335–340. doi: 10.1038/nature18282. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Hussain S., Sajini A.A., Blanco S., et al. NSun2-mediated cytosine-5 methylation of vault noncoding RNA determines its processing into regulatory small RNAs. Cell Rep. 2013;4(2):255–261. doi: 10.1016/j.celrep.2013.06.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Khoddami V., Cairns B.R. Identification of direct targets and modified bases of RNA cytosine methyltransferases. Nat Biotechnol. 2013;31(5):458–464. doi: 10.1038/nbt.2566. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Sajini A.A., Choudhury N.R., Wagner R.E., et al. Loss of 5-methylcytosine alters the biogenesis of vault-derived small RNAs to coordinate epidermal differentiation. Nat Commun. 2019;10:2550. doi: 10.1038/s41467-019-10020-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Botchkarev V.A., Millar S.E. 1st ed. Humana; Totowa, NJ: 2018. Epigenetic Regulation of Skin Development and Regeneration. [Google Scholar]
- 162.Esteve-Puig R., Bueno-Costa A., Writers Esteller M. Readers and erasers of RNA modifications in cancer. Cancer Lett. 2020;474:127–137. doi: 10.1016/j.canlet.2020.01.021. [DOI] [PubMed] [Google Scholar]
- 163.Jonkhout N., Tran J., Smith M.A., et al. The RNA modification landscape in human disease. RNA. 2017;23(12):1754–1769. doi: 10.1261/rna.063503.117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Barrand M.A., Bagrij T., Neo S.Y. Multidrug resistance-associated protein: a protein distinct from P-glycoprotein involved in cytotoxic drug expulsion. Gen Pharmacol. 1997;28(5):639–645. doi: 10.1016/s0306-3623(96)00284-4. [DOI] [PubMed] [Google Scholar]
- 165.Cole S.P., Bhardwaj G., Gerlach J.H., et al. Overexpression of a transporter gene in a multidrug-resistant human lung cancer cell line. Science. 1992;258(5088):1650–1654. doi: 10.1126/science.1360704. [DOI] [PubMed] [Google Scholar]
- 166.Doyle L.A., Yang W., Abruzzo L.V., et al. A multidrug resistance transporter from human MCF-7 breast cancer cells. Proc Natl Acad Sci U S A. 1998;95(26):15665–15670. doi: 10.1073/pnas.95.26.15665. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Roninson I.B. Springer US; Boston, MA: 1991. Molecular and Cellular Biology of Multidrug Resistance in Tumor Cells. [Google Scholar]
- 168.Zaman G.J., Flens M.J., van Leusden M.R., et al. The human multidrug resistance-associated protein MRP is a plasma membrane drug-efflux pump. Proc Natl Acad Sci U S A. 1994;91(19):8822–8826. doi: 10.1073/pnas.91.19.8822. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Borg A.G., Burgess R., Green L.M., et al. Overexpression of lung-resistance protein and increased P-glycoprotein function in acute myeloid leukaemia cells predict a poor response to chemotherapy and reduced patient survival. Br J Haematol. 1998;103(4):1083–1091. doi: 10.1046/j.1365-2141.1998.01111.x. [DOI] [PubMed] [Google Scholar]
- 170.Hu Y., Stephen A.G., Cao J., et al. A very early induction of major vault protein accompanied by increased drug resistance in U-937 cells. Int J Cancer. 2002;97(2):149–156. doi: 10.1002/ijc.1590. [DOI] [PubMed] [Google Scholar]
- 171.Meng C., Wei Z., Zhang Y., et al. Regulation of cytochrome P450 3A4 by small vault RNAb derived from the non-coding vault RNA1 of multidrug resistance-linked vault particle. Mol Med Rep. 2016;14:387–393. doi: 10.3892/mmr.2016.5228. [DOI] [PubMed] [Google Scholar]
- 172.Kitazono M., Okumura H., Ikeda R., et al. Reversal of LRP-associated drug resistance in colon carcinoma SW-620 cells. Int J Cancer. 2001;91:126–131. doi: 10.1002/1097-0215(20010101)91:1<126::aid-ijc1018>3.0.co;2-8. [DOI] [PubMed] [Google Scholar]
- 173.de Wildt S.N., Kearns G.L., Leeder J.S., et al. Cytochrome P450 3A: ontogeny and drug disposition. Clin Pharmacokinet. 1999;37(6):485–505. doi: 10.2165/00003088-199937060-00004. [DOI] [PubMed] [Google Scholar]
- 174.Li J.H., Wang M., Zhang R., et al. E2F1-directed activation of nc886 mediates drug resistance in cervical cancer cells via regulation of major vault protein. Int J Clin Exp Pathol. 2017;10(9):9233–9242. [PMC free article] [PubMed] [Google Scholar]
- 175.Li F., Li J., Wang P.H., et al. SARS-CoV-2 spike promotes inflammation and apoptosis through autophagy by ROS-suppressed PI3K/AKT/mTOR signaling. Biochim Biophys Acta, Mol Basis Dis. 2021;1867(12):166260. doi: 10.1016/j.bbadis.2021.166260. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Li Y., Renner D.M., Comar C.E., et al. SARS-CoV-2 induces double-stranded RNA-mediated innate immune responses in respiratory epithelial-derived cells and cardiomyocytes. Proc Natl Acad Sci U S A. 2021;118(16) doi: 10.1073/pnas.2022643118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Zheng Z.Q., Wang S.Y., Xu Z.S., et al. SARS-CoV-2 nucleocapsid protein impairs stress granule formation to promote viral replication. Cell Discov. 2021;7:38. doi: 10.1038/s41421-021-00275-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Gao B., Gong X., Fang S., et al. Inhibition of anti-viral stress granule formation by coronavirus endoribonuclease nsp 15 ensures efficient virus replication. PLoS Pathog. 2021;17(2) doi: 10.1371/journal.ppat.1008690. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Fort R.S., Mathó C., Geraldo M.V., et al. Nc886 is epigenetically repressed in prostate cancer and acts as a tumor suppressor through the inhibition of cell growth. BMC Cancer. 2018;18:127. doi: 10.1186/s12885-018-4049-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Wan M., Qiu X., Lu Q. High expression of VTRNA2-1 in systemic lupus erythematosus patients' B lymphocytes and its significance. Zhong Nan Da Xue Xue Bao Yi Xue Ban. 2020;45(2):123–127. doi: 10.11817/j.issn.1672-7347.2020.180546. [DOI] [PubMed] [Google Scholar]
- 181.You Y.A., Kwon E.J., Hwang H.S., et al. Elevated methylation of the vault RNA2-1 promoter in maternal blood is associated with preterm birth. BMC Genom. 2021;22:528. doi: 10.1186/s12864-021-07865-y. [DOI] [PMC free article] [PubMed] [Google Scholar]


