Next Article in Journal
Association of the Inactive Circulating Matrix Gla Protein with Vitamin K Intake, Calcification, Mortality, and Cardiovascular Disease: A Review
Next Article in Special Issue
Upregulated gga-miR-16-5p Inhibits the Proliferation Cycle and Promotes the Apoptosis of MG-Infected DF-1 Cells by Repressing PIK3R1-Mediated the PI3K/Akt/NF-κB Pathway to Exert Anti-Inflammatory Effect
Previous Article in Journal
Translational Control of Canonical and Non-Canonical Translation Initiation Factors at the Sea Urchin Egg to Embryo Transition
Previous Article in Special Issue
5p and 3p Strands of miR-34 Family Members Have Differential Effects in Cell Proliferation, Migration, and Invasion in Cervical Cancer Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Implications of the Long Non-Coding RNA NEAT1 in Non-Cancerous Diseases

1
Division of Oncology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria
2
Research Unit of Non-Coding RNAs and Genome Editing in Cancer, Medical University of Graz, 8010 Graz, Austria
3
Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2019, 20(3), 627; https://doi.org/10.3390/ijms20030627
Submission received: 14 December 2018 / Revised: 10 January 2019 / Accepted: 29 January 2019 / Published: 1 February 2019
(This article belongs to the Collection Regulation by Non-coding RNAs)

Abstract

:
Long non-coding RNAs (lncRNAs) are involved in a variety of biological and cellular processes as well as in physiologic and pathophysiologic events. This review summarizes recent literature about the role of the lncRNA nuclear enriched abundant transcript 1 (NEAT1) in non-cancerous diseases with a special focus on viral infections and neurodegenerative diseases. In contrast to its role as competing endogenous RNA (ceRNA) in carcinogenesis, NEAT1’s function in non-cancerous diseases predominantly focuses on paraspeckle-mediated effects on gene expression. This involves processes such as nuclear retention of mRNAs or sequestration of paraspeckle proteins from specific promoters, resulting in transcriptional induction or repression of genes involved in regulating the immune system or neurodegenerative processes. NEAT1 expression is aberrantly—mostly upregulated—in non-cancerous pathological conditions, indicating that it could serve as potential prognostic biomarker. Additional studies are needed to elucidate NEAT1’s capability to be a therapeutic target for non-cancerous diseases.

Graphical Abstract

1. Introduction

According to the project Encyclopedia of DNA Elements (ENCODE), approximately 70% of the human genome is transcribed into RNA but less than 2% is actually protein-coding [1]. Based on this information, it is not surprising that non-coding RNAs (ncRNAs) have increasingly become a strong focus of research at academic centers [2]. Substantial improvements in sequencing technologies have led to the discovery of the large group of ncRNAs. As indicated in the name, ncRNAs are RNA molecules that are not further translated into proteins. The group of ncRNAs can be divided into subgroups, including the approximately 20-nt long microRNAs (miRNAs), [3,4], Piwi-interacting RNAs (piRNAs) [5], small-interfering RNAs (siRNAs) [6], circular RNAs [7], and long non-coding RNAs (lncRNAs) which are typically >200 nt long [8]. While miRNAs and their significance in cellular regulation were discovered in 1993 [9,10], within the past few years, lncRNAs have emerged as interesting molecules [11,12]. Since then, lncRNAs have been found to be involved in a variety of biological processes, including gene expression regulation, subcellular architecture, and protein complex stabilization [13,14], as well as in physiology and pathophysiology [15,16,17]. Within the last few years, numerous studies have shown that the lncRNA nuclear enriched abundant transcript 1 (NEAT1) plays a crucial role in carcinogenesis [18]; emerging evidence, however shows that this lncRNA is also essentially involved in non-cancerous diseases such as neurodegeneration and viral infections. This review aims to provide an overview of information collected thus far pertaining to NEAT1’s role in non-cancer related diseases.

2. NEAT1: Overview, Domain Architecture, Function

The lncRNA NEAT1 is the main actor in this review and is described in detail, ranging from its discovery and architecture to its cellular and physiological functions.

2.1. NEAT1 Overview

Discovered in 2007, NEAT1 is an un-spliced, polyadenylated non-coding transcript with a high abundance in ovary, prostate, colon, and pancreas [19]. The gene encoding for NEAT1 is transcribed by Pol II from the multiple endocrine neoplasia locus (MEN1) in the human chromosome 11q13 [20]. There are two NEAT1 isoforms, i.e., a short 3.7 kb (NEAT1_1) and a long 23 kb version (NEAT1_2) [21]. In contrast to the NEAT1_2 isoform as observed in high abundance in a subpopulation of cells in the stomach and intestine of adult mice, NEAT1_1 exhibits a high expression in a wide range of tissues. Accordingly, NEAT1_2-dependent paraspeckle formation is primarily detectable in cellular subpopulations of mice [22]. NEAT1 is—as indicated in the name—enriched in the nucleus and has been shown to function as an essential structural component of paraspeckles and determines the integrity of these subnuclear bodies [19,23]. Cellular depletion of NEAT1 results in a loss of paraspeckles. Overexpression of NEAT1—but not of the paraspeckle component 1 (PSPC1) protein—leads to an increased paraspeckle accumulation pointing towards NEAT1 being the bottleneck of paraspeckle formation [23]. Paraspeckles are subnuclear ribonucleoprotein bodies composed of the lncRNA NEAT1 and the core proteins polypyrimidine tract-binding protein PTB-associated splicing factor/splicing factor proline glutamine rich (PSF/SFPQ) [24], 54 kDa nuclear RNA- and DNA-binding protein/non-POU domain-containing octamer-binding protein (p54nrb/NONO) and PSPC1 [25,26]. Paraspeckles are involved in regulating gene expression by a process called nuclear retention. Adenosine-to-inosine (A–I) edited mRNAs are retained in the nucleus whereas unedited mRNAs are transported into the cytoplasm [27,28]. Adenosine-to-inosine editing is a nuclear process which is catalyzed by dsRNA-dependent adenosine deaminases (ADARs) leading to a hydrolytic deamination of adenosine to inosine in double-stranded regions of targeted mRNAs. Adenosine-to-inosine editing predominantly occurs in inverted repeated Alu elements (IRAlus) [28,29]. Paraspeckles can therefore be seen as nuclear mRNA anchors [30].

2.2. NEAT1 Domain Architecture

Two of the paraspeckle protein components (p54nrb/NONO and PSPC1) were found to form heterodimers within paraspeckles [31] and exhibit an intensive co-localization with NEAT1 [23]. Three protein interaction sites located near the 5’ and 3’ ends of NEAT1 are a prerequisite in order to bind to p54nrb/NONO [32] (Figure 1A). An advanced combination of protein and RNA visualization techniques, namely structured illumination microscopy (SIM) and fluorescent in situ hybridization (FISH), allowed for the simultaneous detection of NEAT1 and the protein components within paraspeckles [33]. NEAT1 has been shown to arrange itself in a core-shell spheroidal structure. The 5’ and 3’ ends of NEAT1 are located at the periphery of the speckles whereas the central sequence is localized within the core [34] (Figure 1B).
Both NEAT1’s middle domain and the binding of p54nrb/NONO to this central site are two features essential and sufficient for paraspeckle formation [35]. Li et al. [36] provided data showing that only the long 23 kb NEAT1 isoform is a major and essential component of paraspeckles, suggesting that the short isoform might be implicated in other cellular functions. Lin et al. [37] provided a structural model of the long NEAT1 isoform showing that long-range interactions between its 5’ and 3’ ends may be important for its architectural role within paraspeckles. Several studies demonstrate that the structure of lncRNAs is defining their function. LncRNAs tend to acquire complex secondary and tertiary structures and it was observed that structural conservation rather than nucleotide sequence conservation is crucial for maintaining their function [38]. In addition to its domains relevant for paraspeckle assembly, NEAT1’s domain architecture and interaction with other proteins is also regulating global pri-miRNA processing. As described above, NEAT1 interacts with p54nrb/NONO and PSF/SFPQ as well as with other RNA-binding proteins. Furthermore, it possesses multiple RNA segments including a “pseudo pri-miRNA” near the 3’ end, which—together with the above mentioned protein-NEAT1 interactions—help to attract the microprocessor Drosha/DiGeorge syndrome critical region 8 (DGCR8) complex responsible for pri-miRNA processing [39].

2.3. Cellular Function of NEAT1

In the cellular context, NEAT1_2 is responsible for the sequestration of paraspeckle components in the drosophila behavior human splicing (DBHS) family, i.e., PSF/SFPQ, PSPC1, and p54nrb/NONO. PSPC1 and p54nrb/NONO regulate the A–I editing of mRNAs [23] and additionally, p54nrb/NONO is involved in retaining those edited mRNAs, preventing their nuclear export [40]. NEAT1 itself is retained in the nucleus without being A–I edited [23]. Depending on the presence of NEAT1 and the associated abundance of paraspeckles in the nucleus, gene expression regulation is influenced to a variable extent. In the case of low paraspeckle accumulation within the nucleus, the high concentration of unbound paraspeckle protein components PSPC1, PSF/SFPQ, and p54nrb/NONO, results in an increased transcriptional regulation of specific genes through their action as positive or negative transcription regulators. Furthermore, a low amount of paraspeckles leads to a decreased nuclear retention of A–I edited mRNAs, thus leaving the export of those mRNAs to the cytoplasm uninfluenced. Conversely, increased paraspeckle formation means a lower concentration of unbound paraspeckle components, therefore limiting their effect on the transcriptional regulation. In addition, A–I edited mRNAs are more efficiently bound by paraspeckles and in turn more effectively retained in the nucleus instead of being transported to the cytoplasm [41] (Figure 2).
Another mode of action of NEAT1 is sponging of miRNAs, which is a function already well documented in carcinogenesis [18] and described in Section 2.5.

2.4. NEAT1 in Physiology

Nakagawa et al. [22] propose that paraspeckles are non-essential, cell subpopulation specific nuclear bodies, since NEAT1 knock-out mice are viable, fertile, and do not show an apparent phenotype. In a subsequent study, the same group provided data that NEAT1_2-mediated paraspeckles are fundamental for corpus luteum formation and contradictory to the earlier results partially define fertility in a subpopulation of mice [42]. Furthermore, NEAT1 was shown to be essential for mammary gland development and the lactation capacity in mice [43] as well as modulating neuronal excitability in humans [44]. As Chen and Carmichael [28] demonstrated, NEAT1_2 is also involved in the differentiation of human embryonic stem cells (hESC). Although hESC mRNAs contain IRAlu elements, and thus, are likely to be A–I edited, these mRNAs are not retained in the nucleus but are transported into the cytoplasm. They are subjected to nuclear retention only after differentiation and the reason why can be found when looking at the difference between NEAT1_2 expression levels. While undifferentiated hESC lack NEAT1_2-dependent paraspeckles (which, in turn, regulate nuclear retention), NEAT1_2 expression is initiated by differentiating enabling the paraspeckle formation and subsequent nuclear retention of hESC mRNAs [28]. NEAT1_2 serves an additional functional role in the transcriptional regulation of Interleukin-8 (IL-8). In a viral infection, NEAT1 sequesters the IL-8 repressor PSF/SFPQ from the IL-8 promoter to paraspeckles. As a result, IL-8 transcription is initiated, leading to immune response stimulation [41].

2.5. NEAT1 in Carcinogenesis

Meta-analyses demonstrate an intensive upregulation of NEAT1 in several cancer entities, resulting in an unfavorable outcome as well as a drastic decrease in overall survival, suggesting a potential role for NEAT1 as prognostic biomarker [45,46]. Numerous studies focusing on NEAT1’s role in cancer biology indicate that this lncRNA is a crucial part of carcinogenesis as found in non-small lung cancer [47,48,49,50], breast cancer [51,52,53], hepatocellular carcinoma [54,55,56,57], ovarian cancer [58,59,60,61], and prostate cancer [62,63], just to name a few. In terms of carcinogenesis, NEAT1 mainly functions as competing endogenous RNA (ceRNA) by sponging tumor-suppressive miRNAs [64]. Subsequently, these miRNAs lose the ability to function as a tumor suppressor and oncogenic mRNAs are translated, ultimately contributing to tumorigenesis [65]. As this review focuses on NEAT1 in non-cancerous diseases, we will refrain from providing more details. For a recent overview of NEAT1’s role in carcinogenesis, please see Klec et al. [18].

3. Immune System and Viral Diseases

Over the past ten years, miRNAs and lncRNAs have become key players in immune system response as well as cancer immunotherapy [66,67]. LncRNAs have been demonstrated to be expressed in a lineage-specific manner, for example, in T-cell population subsets [68] or in a certain subset of lymphocytes [69]. Furthermore, lncRNAs were shown to be involved in controlling the differentiation and function of innate and adaptive immune cell types. Examples of lncRNAs in immune system regulation include: (1) the lncRNA H19 regulates hematopoietic development [70], (2) Morrbid and lnc-DC are involved in the regulation of myeloid cell survival and myeloid cell differentiation [71,72], (3) lincR-Ccr2-5’AS controls CD4+ T-cell differentiation [73], and (4). lincRNA-Cox2 is an activator of inflammation [74]. Chen et al. [75] provided a detailed overview of lncRNAs in immune system regulation.
Concerning NEAT1, its involvement in immune system responses was initially discovered in the brains of mice infected with either Japanese encephalitis virus or the rabies virus [13,76]. Since this discovery, NEAT1’s role in regulating the immune system has been studied intensively and is summarized in the next section and in Table 1.

3.1. Sepsis and Sepsis-Induced Acute Kidney Injury

Non-coding RNAs have been shown to play an important role in the host defense system and viral infections [77,78]. NEAT1 was shown to be differentially expressed and is proposed to be a suitable candidate as an additive biomarker for early sepsis detection as it was found to be highly upregulated in peripheral blood mononuclear cells (PBMCs) in sepsis patients compared to healthy controls [79], and in sepsis-induced acute kidney injury (AKI) [80]. In the case of sepsis-induced AKI, NEAT1 expression correlates positively with the severity of the disease. Knock-down of NEAT1 in rat kidney cells leads to reduced lipopolysaccharide (LPS)-induced cell injury by an accompanied upregulation of miR-204. miR-204 has been shown to protect the kidney during sepsis by regulating Hmx1 (heme oxygenase) [81]. These protective functions are lost when increased NEAT1 levels sponge miR-204, activating NF-κB signaling and leading to sepsis associated organ failure [82] in sepsis-induced AKI patients [80].

3.2. Viral-Induced Diseases

As a stress-induced lncRNA, NEAT1 expression increases in response to a viral infection. Over the last two years, numerous viral diseases have been correlated with differences in NEAT1 expression, either acting anti-viral or pro-viral.

3.2.1. Anti-viral Effects of NEAT1

NEAT1 was first linked to an infection with the human immunodeficiency virus (HIV-1) in 2013. Zhang’s group [83] detected an increased NEAT1 expression after HIV-1 infection. A new finding at that time, the paraspeckle components PSF/SFPQ, p54nrb/NONO, and matrin 3 had until then only been associated with HIV infection. Knockdown of NEAT1 in the T-cell lines Jurkat and MT4 resulted in an increased HIV-1 production due to a more pronounced nucleus-to-cytoplasm export of Rev-dependent instability elements (INS)-containing HIV-1 mRNA, or in other words, decreased paraspeckle-mediated nuclear retention [83]. A subsequent study could shed more light on how the underlying mechanism is regulated. Exportin 1 (XPO1) is the Rev-dependent mediator of the abovementioned nuclear export. The nucleus-to-cytoplasm transfer of these incompletely spliced viral transcripts regulates gene expression post-transcriptionally. The export of these un-spliced transcripts is inhibited due to inefficient splicing and the contained INS elements. As paraspeckles depend crucially on NEAT1 and have been shown to retain these INS-containing transcripts in the nucleus, it was proposed that increased HIV-1 replication after NEAT1 knock-down is a consequence of decreased nuclear retention of HIV-1 mRNA [84]. Pandey et al. [85] reports on differential NEAT1 expression in dengue disease, proposing that NEAT1 could be a marker for the progression of dengue fever since its expression is reduced in severe phenotypes found in dengue disease. Interestingly, NEAT1 expression was highest in the early stages of the disease, decreasing with progression [85]. The authors suggest that low NEAT1 expression could induce apoptosis via p53 [86] in monocytes, as already reported in myeloid lineage cells [76] or breast cancer cell lines [51]. Hantavirus infection is followed by increased NEAT1 expression which, on the one hand, controls viral replication and induces anti-viral immune response by promoting interferon (IFN) production via retinoic acid inducible gene I (RIG-I) signaling, on the other hand. The mechanism behind is the above described SFPQ sequestration to paraspeckles away from the RIG-I promoter which is known to be a positive regulator of IFN-gene activation [87], therefore, promoting anti-viral immunity [88,89]. Beeharry et al. [90] demonstrated that the Hepatitis D virus (HDV) interacts with the major paraspeckle components, i.e., PSF/SFPQ, PSPC1, and p54nrb/NONO, indicating a crucial role of paraspeckles in HDV infection. Indeed, viral replication was shown to depend crucially on this interaction as a knockdown of these proteins leads to a hampered HDV replication. Upon HDV infection, NEAT1 levels are upregulated and NEAT1 foci are enlarged. Due to the facts that IL-8 levels are 2-fold increased upon HDV infection and a knockdown of paraspeckle proteins results in reduced HDV replication, it is tempting to speculate that NEAT1 upregulation-induced sequestration of paraspeckle proteins is causing the anti-viral effects leading to the activation of innate immunity [90,91]. The upregulation of IL-8 is based on the NEAT1-induced relocation of the paraspeckle component SFPQ to paraspeckles where it is unable to execute its repressor function on IL-8 transcription [91].

3.2.2. Pro-viral Effects of NEAT1

Recently, two independent groups provided data on NEAT1 upregulation after an infection with the herpes simplex virus (HSV) [92,93]. Viollet et al. [92] demonstrated that approximately 210 genes are upregulated after Kaposi sarcoma-associated herpesvirus (KSHV) infection, with NEAT1 showing a 3-fold increase in expression in KSHV infected cells versus non-infected cells under hypoxic conditions. The hypoxia-inducible factor 2 (HIF2) is a known regulator of NEAT1 transcription [94]. Since NEAT1 has been demonstrated to increase survival of cancer cells [18,94], the authors propose that an upregulation of HIF-responsive genes causes tumorigenesis leading to the development of Kaposi sarcoma and other KSHV-induced tumors [92]. Wang et al. [93] showed that NEAT1 is upregulated after a herpes simplex infection in a STAT3-dependent manner. The HSV-1 genome is recruited to paraspeckles to regulate its transcription. NEAT1 upregulation facilitates virus replication by mediating the interaction between the paraspeckle components p54nrb/NONO, and PSPC1 and herpes simplex gene promoters, ultimately leading to an increased viral gene expression. Knockdown of the anti-viral SFPQ results in increased replication due to facilitation of the interaction between STAT3 and viral gene promoters. Initial advances pertaining to therapeutic interventions by NEAT1 modulation show that in the case of herpes simplex infection, thermosensitive gels coated with siRNA against NEAT1 were able to reduce virus induced skin lesions [93]. These data show on the one hand that HSV-1 replication is regulated by a NEAT1-dependent paraspeckle-mediated transcriptional cascade and on the other hand that NEAT1 upregulation seems to be a general response to viral infections. If the consequences are pro-viral or anti-viral depends on the downstream mechanisms.

4. Neurodegeneration and Neuronal Defects

Numerous studies show that lncRNAs are involved in regulating and/or protecting against neurodegeneration [95]. There are several indications underlining the importance of lncRNAs in brain function and the development of neurodegenerative diseases: (1) the existence of brain-specific lncRNAs with precisely regulated temporal and spatial expression patterns [96]; (2) the correlation between highly transcriptional active CNS cells and the fact that lncRNAs are involved in transcriptional regulation; (3) the tissue-specific expression of certain lncRNAs either in particular regions of the CNS or even in different cell types [97,98]; and (4) the observation that dysregulations or mutations in lncRNA gene loci are associated with neurodegenerative disorders [99]. Several lncRNAs have been demonstrated to be involved in the development and progression of neurodegenerative diseases. Some well-studied examples include the following: (1) the β-site amyloid β-protein precursor (APP) cleaving enzyme 1 antisense RNA (BACE1-AS) in Alzheimer’s disease (AD); (2) phosphatase and tensin homolog (PTEN)-induced kinase 1 antisense (PINK1-AS) which is stabilizing its protein-coding pendant PINK1 in Parkinson’s disease (PD) [100,101,102]; and (3) brain-derived neurotrophic factor antisense (BDNF-AS) in Huntington’s disease (HD) [103,104]. More detailed information about the role of lncRNAs in neurodegenerative diseases are well reviewed by Quan et al. [99] and Wan et al. [105].
NEAT1 is speculated to have biological functions in the brain’s pathologies since the expression level of this lncRNA increases significantly in the nucleus accumbens of heroin users [106] and is described as a mediator of the neuroprotective effects of bexarotene on traumatic brain injury in mice [107]. Table 2 summarizes the neurodegenerative diseases with NEAT1 contribution, which are also discussed in detail below.

4.1. Huntington’s Disease

HD is caused by an expansion of a CAG triplet repeat within the huntingtin gene, creating a mutant version of the huntingtin protein [108]. There is some controversy as to whether or not NEAT1 actively contributes to Huntington’s disease pathogenesis or if it triggers neuroprotective mechanisms. Microarray analyses as well as quantitative PCR of post-mortem brains of Huntington’s disease patients and R6/2 mouse brains—a model for HD—show an upregulation of NEAT1 expression. Overexpression of NEAT1 in neuro2A cells increases cell viability upon neuronal injury induced by H2O2 treatment, thus the authors suggest a protective mechanism against oxidative injury in HD pathogenesis rather than a contribution to disease development [109]. A review by Johnson et al. [110] reports contrary findings. Chip sequencing indicates that NEAT1 is a target of REST (a transcriptional repressor) and p53 (a tumor suppressor), both of which are known to be key players in HD. Changes in lncRNA expression are considered to result in altered epigenetic gene regulation in diseased neurons, and thus are believed to be possible contributors to HD pathology [110]. Further studies are needed to unravel NEAT1’s role in HD pathogenesis.

4.2. Multiple Sclerosis

Multiple sclerosis (MS) is a chronic autoimmune, inflammatory neurological disease of the central nervous system (CNS) [111]. A screening of 84 lncRNAs revealed an upregulation of NEAT1 in the serum of patients suffering from multiple sclerosis (MS) compared to either healthy controls or patients with who have idiopathic inflammatory myopathy (IIM) [112]. Taking into consideration that NEAT1 plays a role in immunity by driving IL-8 activation (in addition to findings by Lund et al. [113] which demonstrate that IL-8 levels increase significantly in MS patients), the authors speculate that upregulated NEAT1 expression activates IL-8 transcription. In addition to IL-8 upregulation, a co-localization of stathmin and toll-like receptor 3 (TLR3) has been found in astrocytes, microglia, and neurons in the brains of MS patients [114], indicating that these players contribute to MS pathogenesis [112].

4.3. Amyotrophic Lateral Sclerosis

Amyotrophic lateral sclerosis (ALS) is a fatal motor neuron disorder which is characterized by progressive loss of the upper and lower motor neurons [115]. NEAT1 is upregulated in ALS patients in the early stages of the disease. Nishimoto et al. [116] were able to show a direct interaction of NEAT1 with two RNA-binding proteins, both of which are mutated in ALS patients, i.e., TAR DNA-binding protein 43 (TDP-43) [117] and fused in sarcoma/translocated in liposarcoma (FUS/TLS) [118]. Both proteins were shown to be enriched in paraspeckles of cultured cells. Interestingly, NEAT1 expression was absent in motor neurons in the spinal cords of healthy control mice but there was a high density of NEAT1 as well as paraspeckles in motor neurons in the spinal cords of ALS patients in the early phase of disease. Based on these results, the authors claim that paraspeckles directly contribute to neurodegenerative diseases [116].

4.4. Parkinson’s Disease

Parkinson’s disease (PD) is a chronic, progressive movement disorder due to a loss of dopamine producing cells in the brain [119]. NEAT1 is upregulated in a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD mouse model together with PINK1 (a known contributor to PD [120]) which was stabilized by NEAT1. Since a knockdown of NEAT1 leads to reduced MPTP-initiated autophagy in vivo resulting in a decreased neuronal injury, authors claim that there is a contribution of NEAT1 in PD. [121] Nearly simultaneously, Liu’s group [122] published supportive results by also showing an upregulation of NEAT1 in an MPTP-induced PD mouse model as well as in a 1-methyl-4-phenylpyridinium (MPP+)-induced PD cell line. NEAT1 knockdown resulted in an increased viability, inhibition of apoptosis, and decreased α-synuclein expression in the PD cell model. Since α-synuclein overexpression reversed the effects of NEAT1 knockdown, a protective role of NEAT1 downregulation in the MPTP-induced PD mouse model was suggested [122].

5. Conclusions

Summarizing recent data pertaining to NEAT1, one can draw the conclusion that this lncRNA contributes to regulating viral diseases and neurodegeneration. One shared feature of the so far investigated non-cancerous diseases is a common upregulation of NEAT1 which highlights the enormous potential of this lncRNA as diagnostic biomarker in these pathologies. Although increased NEAT1 levels seem to be a common event upon viral infection and in neurodegenerative diseases, the consequences of NEAT1 upregulation are diverse. We hypothesize that the mode of NEAT1’s action—if it acts pro-viral or anti-viral or if it is contributing to neurodegeneration or protecting from it—depends on the occurring downstream events. From the mechanistic point of view, NEAT1 either exerts the function of paraspeckle-mediated nuclear retention (as in HIV infection), transcriptional regulation by sequestering paraspeckle proteins (as in HTNV, HDV and Herpes simplex infections as well as in Multiple Sclerosis) or sponging of miRNAs (as in sepsis-induced AKI). Concerning the other discussed diseases more studies are needed to investigate the consequences of NEAT1 upregulation and the underlying mechanisms. Although nuclear retention and transcriptional regulation by sequestration of paraspeckle proteins seem to be NEAT1’s predominant modes of action in non-cancerous diseases, there is emerging evidence that it also can act as sponge for miRNAs. For instance, Wang et al. [123] observed that, in the context of diabetic nephropathy (DN) progression, NEAT1 directly binds to miR-27b-3p, leading to the suppression of its function. As miR-27b-3p directly targets ZEB1 (a key player in the EMT process), the group suggests that inhibition of NEAT1 represses DN progression through regulating EMT (and fibrogenesis). Further evidence which consolidates NEAT1’s ability to sponge miRNAs was provided by Wang et al. [124] by demonstrating that NEAT1 directly binds to miR-342-3p in the context of atherosclerotic cardiovascular diseases. Wang’s group observed that a knockdown of NEAT1 represses the inflammation response and inhibits lipid uptake by THP-1 cells in a miR-342-3p-dependent manner. Chen et al. [125] corroborate NEAT1’s ability to sponge miRNAs in the context of atherosclerosis. Their group observed that NEAT1 directly binds to miR-128 and through that interaction plays a role in oxidized low density lipoprotein (ox-LDL)-induced inflammation and oxidative stress in atherosclerosis development. Despite the fact that there are reports on NEAT1 influencing disease progression by sponging miRNAs, the described paraspeckle-mediated effects on the transcriptional regulation seem to be of greater significance in the context of non-cancerous diseases. NEAT1’s suitability as therapeutic target still needs more intensive research. As already mentioned above, first advances towards a therapeutic application of NEAT1 have been made in the context of Herpes simplex infection where virus-induced skin lesions have successfully been treated with gels containing NEAT1 siRNA [93]. Therefore, we believe that NEAT1 plays a crucial role in non-cancerous diseases and future studies will help to understand the complete story of this interesting lncRNA.

Author Contributions

All authors contributed to drafting, writing, and reviewing of this article.

Acknowledgments

No funding was used for writing this manuscript. We greatly thank Nadine Lichtenberger for proofreading and editing our manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

A–Iadenosine-to-inosine
ADAlzheimer’s disease
ADARsdsRNA dependent adenosine deaminases
AKIacute kidney injury
ALSamyotrophic lateral sclerosis
APPamyloid-β protein precursor
BACE1-ASβ-site APP cleaving enzyme 1 antisense RNA
BDNFbrain derived neurotrophic factor
ceRNAcompeting endogenous RNA
CNScentral nervous system
DBHSdrosophila behavior human splicing
DGCR8DiGeorge syndrome critical region 8
FISHfluorescence in situ hybridization
FUS/TLSfused in sarcoma/translocated in liposarcoma
HDHuntington’s disease
HDVhepatitis D virus
hESChuman embryonic stem cells
HIV-1human immunodeficiency virus 1
Hmx1heme oxygenase 1
HTNVhantavirus
IFNinterferon
IIMidiopathic inflammatory myopathy
INS elementsinstability elements
IRAluinverted repeated Alu elements
KSHVKaposi Sarcoma-Associated Herpesvirus
LPSlipopolysaccharide
(l)ncRNA(long) non-coding RNA
miRNAmicroRNA
MPP+1-methyl-4-phenylpyridinium
MPTP1-methyl-4-phenyl-1,2,3,6-tetrahydropyridin
mRNAmessenger RNA
MSmultiple sclerosis
NEAT1nuclear enriched abundant transcript 1
ox-LDLoxidizied low density lipoprotein
p54nrb/NONO54 kDa nuclear RNA- and DNA-binding protein/Non-POU domain-containing octamer-binding protein
PBMCperipheral blood mononuclear cells
PDParkinson’s disease
PINK1PTEN-induced kinase 1
piRNApiwi-interacting RNA
Pol IIRNA polymerase II
PSF/SFPQPTB-associated splicing protein/splicing factor proline glutamine rich
PSPC1paraspeckle component 1
PTBpolypyrimidine tract-binding protein
PTENphosphatase and tensin homolog
RIG-Iretinoic acid inducible gene I
SIMstructure illumination microscopy
siRNAshort interfering RNA
TDP-43TAR DNA-binding protein 43
TLR3toll-like receptor 3
XPO1exportin 1

References

  1. Consortium, E.P. An integrated encyclopedia of DNA elements in the human genome. Nature 2012, 489, 57–74. [Google Scholar] [CrossRef] [PubMed]
  2. Ling, H.; Vincent, K.; Pichler, M.; Fodde, R.; Berindan-Neagoe, I.; Slack, F.J.; Calin, G.A. Junk DNA and the long non-coding RNA twist in cancer genetics. Oncogene 2015, 34, 5003–5011. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Wightman, B.; Ha, I.; Ruvkun, G. Posttranscriptional regulation of the heterochronic gene lin-14 by lin-4 mediates temporal pattern formation in C. elegans. Cell 1993, 75, 855–862. [Google Scholar] [CrossRef]
  4. Lee, R.C.; Feinbaum, R.L.; Ambros, V. The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell 1993, 75, 843–854. [Google Scholar] [CrossRef]
  5. Vagin, V.V.; Sigova, A.; Li, C.; Seitz, H.; Gvozdev, V.; Zamore, P.D. A distinct small RNA pathway silences selfish genetic elements in the germline. Science 2006, 313, 320–324. [Google Scholar] [CrossRef] [PubMed]
  6. Mello, C.C.; Conte, D., Jr. Revealing the world of RNA interference. Nature 2004, 431, 338–342. [Google Scholar] [CrossRef] [PubMed]
  7. Salzman, J.; Gawad, C.; Wang, P.L.; Lacayo, N.; Brown, P.O. Circular RNAs are the predominant transcript isoform from hundreds of human genes in diverse cell types. PLoS ONE 2012, 7, e30733. [Google Scholar] [CrossRef]
  8. Mercer, T.R.; Dinger, M.E.; Mattick, J.S. Long non-coding RNAs: Insights into functions. Nat. Rev. Genet. 2009, 10, 155–159. [Google Scholar] [CrossRef]
  9. Stiegelbauer, V.; Vychytilova-Faltejskova, P.; Karbiener, M.; Pehserl, A.M.; Reicher, A.; Resel, M.; Heitzer, E.; Ivan, C.; Bullock, M.; Ling, H.; et al. miR-196b-5p Regulates Colorectal Cancer Cell Migration and Metastases through Interaction with HOXB7 and GALNT5. Clin. Cancer Res. 2017, 23, 5255–5266. [Google Scholar] [CrossRef]
  10. Pichler, M.; Calin, G.A. MicroRNAs in cancer: From developmental genes in worms to their clinical application in patients. Br. J. Cancer 2015, 113, 569–573. [Google Scholar] [CrossRef]
  11. Fosselteder, J.; Calin, G.A.; Pichler, M. Long non-coding RNA CCAT2 as a therapeutic target in colorectal cancer. Expert Opin. Targets 2018, 22, 973–976. [Google Scholar] [CrossRef] [PubMed]
  12. Rigoutsos, I.; Lee, S.K.; Nam, S.Y.; Anfossi, S.; Pasculli, B.; Pichler, M.; Jing, Y.; Rodriguez-Aguayo, C.; Telonis, A.G.; Rossi, S.; et al. N-BLR, a primate-specific non-coding transcript leads to colorectal cancer invasion and migration. Genome Biol. 2017, 18, 98. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Kung, J.T.; Colognori, D.; Lee, J.T. Long noncoding RNAs: Past, present, and future. Genetics 2013, 193, 651–669. [Google Scholar] [CrossRef] [PubMed]
  14. Gutschner, T.; Richtig, G.; Haemmerle, M.; Pichler, M. From biomarkers to therapeutic targets-the promises and perils of long non-coding RNAs in cancer. Cancer Metastasis Rev. 2018, 37, 83–105. [Google Scholar] [CrossRef] [PubMed]
  15. Smolle, M.A.; Bauernhofer, T.; Pummer, K.; Calin, G.A.; Pichler, M. Current Insights into Long Non-Coding RNAs (LncRNAs) in Prostate Cancer. Int. J. Mol. Sci. 2017, 18, 473. [Google Scholar] [CrossRef] [PubMed]
  16. Del Vecchio, F.; Lee, G.H.; Hawezi, J.; Bhome, R.; Pugh, S.; Sayan, E.; Thomas, G.; Packham, G.; Primrose, J.; Pichler, M.; et al. Long non-coding RNAs within the tumour microenvironment and their role in tumour-stroma cross-talk. Cancer Lett. 2018, 421, 94–102. [Google Scholar] [CrossRef] [PubMed]
  17. Smolle, M.A.; Pichler, M. The Role of Long Non-Coding RNAs in Osteosarcoma. Non-Coding RNA 2018, 4, 7. [Google Scholar] [CrossRef] [PubMed]
  18. Klec, C.; Prinz, F.; Pichler, M. Involvement of the long non-coding RNA NEAT1 in carcinogenesis. Mol. Oncol. 2018. [Google Scholar] [CrossRef]
  19. Hutchinson, J.N.; Ensminger, A.W.; Clemson, C.M.; Lynch, C.R.; Lawrence, J.B.; Chess, A. A screen for nuclear transcripts identifies two linked noncoding RNAs associated with SC35 splicing domains. BMC Genom. 2007, 8, 39. [Google Scholar] [CrossRef] [PubMed]
  20. Guru, S.C.; Agarwal, S.K.; Manickam, P.; Olufemi, S.E.; Crabtree, J.S.; Weisemann, J.M.; Kester, M.B.; Kim, Y.S.; Wang, Y.; Emmert-Buck, M.R.; et al. A transcript map for the 2.8-Mb region containing the multiple endocrine neoplasia type 1 locus. Genome Res. 1997, 7, 725–735. [Google Scholar] [CrossRef] [PubMed]
  21. Sasaki, Y.T.; Ideue, T.; Sano, M.; Mituyama, T.; Hirose, T. MENepsilon/beta noncoding RNAs are essential for structural integrity of nuclear paraspeckles. Proc. Natl. Acad. Sci. USA 2009, 106, 2525–2530. [Google Scholar] [CrossRef] [PubMed]
  22. Nakagawa, S.; Naganuma, T.; Shioi, G.; Hirose, T. Paraspeckles are subpopulation-specific nuclear bodies that are not essential in mice. J. Cell Biol. 2011, 193, 31–39. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Clemson, C.M.; Hutchinson, J.N.; Sara, S.A.; Ensminger, A.W.; Fox, A.H.; Chess, A.; Lawrence, J.B. An architectural role for a nuclear noncoding RNA: NEAT1 RNA is essential for the structure of paraspeckles. Mol. Cell 2009, 33, 717–726. [Google Scholar] [CrossRef] [PubMed]
  24. Prasanth, K.V.; Prasanth, S.G.; Xuan, Z.; Hearn, S.; Freier, S.M.; Bennett, C.F.; Zhang, M.Q.; Spector, D.L. Regulating gene expression through RNA nuclear retention. Cell 2005, 123, 249–263. [Google Scholar] [CrossRef] [PubMed]
  25. Fox, A.H.; Lam, Y.W.; Leung, A.K.; Lyon, C.E.; Andersen, J.; Mann, M.; Lamond, A.I. Paraspeckles: A novel nuclear domain. Curr. Biol. 2002, 12, 13–25. [Google Scholar] [CrossRef]
  26. Bond, C.S.; Fox, A.H. Paraspeckles: Nuclear bodies built on long noncoding RNA. J. Cell Biol. 2009, 186, 637–644. [Google Scholar] [CrossRef]
  27. Zhang, Z.; Carmichael, G.G. The fate of dsRNA in the nucleus: A p54(nrb)-containing complex mediates the nuclear retention of promiscuously A-to-I edited RNAs. Cell 2001, 106, 465–475. [Google Scholar] [CrossRef]
  28. Chen, L.L.; Carmichael, G.G. Altered nuclear retention of mRNAs containing inverted repeats in human embryonic stem cells: Functional role of a nuclear noncoding RNA. Mol. Cell 2009, 35, 467–478. [Google Scholar] [CrossRef]
  29. Bass, B.L. RNA editing by adenosine deaminases that act on RNA. Annu. Rev. Biochem. 2002, 71, 817–846. [Google Scholar] [CrossRef]
  30. Torres, M.; Becquet, D.; Blanchard, M.P.; Guillen, S.; Boyer, B.; Moreno, M.; Franc, J.L.; Francois-Bellan, A.M. Paraspeckles as rhythmic nuclear mRNA anchorages responsible for circadian gene expression. Nucleus 2017, 8, 249–254. [Google Scholar] [CrossRef]
  31. Fox, A.H.; Bond, C.S.; Lamond, A.I. P54nrb forms a heterodimer with PSP1 that localizes to paraspeckles in an RNA-dependent manner. Mol. Biol. Cell 2005, 16, 5304–5315. [Google Scholar] [CrossRef] [PubMed]
  32. Murthy, U.M.; Rangarajan, P.N. Identification of protein interaction regions of VINC/NEAT1/Men epsilon RNA. FEBS Lett. 2010, 584, 1531–1535. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Mito, M.; Kawaguchi, T.; Hirose, T.; Nakagawa, S. Simultaneous multicolor detection of RNA and proteins using super-resolution microscopy. Methods 2016, 98, 158–165. [Google Scholar] [CrossRef] [PubMed]
  34. West, J.A.; Mito, M.; Kurosaka, S.; Takumi, T.; Tanegashima, C.; Chujo, T.; Yanaka, K.; Kingston, R.E.; Hirose, T.; Bond, C.; et al. Structural, super-resolution microscopy analysis of paraspeckle nuclear body organization. J. Cell Biol. 2016, 214, 817–830. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Yamazaki, T.; Souquere, S.; Chujo, T.; Kobelke, S.; Chong, Y.S.; Fox, A.H.; Bond, C.S.; Nakagawa, S.; Pierron, G.; Hirose, T. Functional Domains of NEAT1 Architectural lncRNA Induce Paraspeckle Assembly through Phase Separation. Mol. Cell 2018. [Google Scholar] [CrossRef] [PubMed]
  36. Li, R.; Harvey, A.R.; Hodgetts, S.I.; Fox, A.H. Functional dissection of NEAT1 using genome editing reveals substantial localization of the NEAT1_1 isoform outside paraspeckles. RNA 2017, 23, 872–881. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Lin, Y.; Schmidt, B.F.; Bruchez, M.P.; McManus, C.J. Structural analyses of NEAT1 lncRNAs suggest long-range RNA interactions that may contribute to paraspeckle architecture. Nucleic Acids Res. 2018, 46, 3742–3752. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Zampetaki, A.; Albrecht, A.; Steinhofel, K. Long Non-coding RNA Structure and Function: Is There a Link? Front. Physiol. 2018, 9, 1201. [Google Scholar] [CrossRef] [PubMed]
  39. Jiang, L.; Shao, C.; Wu, Q.J.; Chen, G.; Zhou, J.; Yang, B.; Li, H.; Gou, L.T.; Zhang, Y.; Wang, Y.; et al. NEAT1 scaffolds RNA-binding proteins and the Microprocessor to globally enhance pri-miRNA processing. Nat. Struct. Mol. Biol. 2017, 24, 816–824. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  40. Chen, L.L.; DeCerbo, J.N.; Carmichael, G.G. Alu element-mediated gene silencing. EMBO J. 2008, 27, 1694–1705. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  41. Hirose, T.; Virnicchi, G.; Tanigawa, A.; Naganuma, T.; Li, R.; Kimura, H.; Yokoi, T.; Nakagawa, S.; Benard, M.; Fox, A.H.; et al. NEAT1 long noncoding RNA regulates transcription via protein sequestration within subnuclear bodies. Mol. Biol. Cell 2014, 25, 169–183. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Nakagawa, S.; Shimada, M.; Yanaka, K.; Mito, M.; Arai, T.; Takahashi, E.; Fujita, Y.; Fujimori, T.; Standaert, L.; Marine, J.C.; et al. The lncRNA Neat1 is required for corpus luteum formation and the establishment of pregnancy in a subpopulation of mice. Development 2014, 141, 4618–4627. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Standaert, L.; Adriaens, C.; Radaelli, E.; Van Keymeulen, A.; Blanpain, C.; Hirose, T.; Nakagawa, S.; Marine, J.C. The long noncoding RNA Neat1 is required for mammary gland development and lactation. RNA 2014, 20, 1844–1849. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Barry, G.; Briggs, J.A.; Hwang, D.W.; Nayler, S.P.; Fortuna, P.R.; Jonkhout, N.; Dachet, F.; Maag, J.L.; Mestdagh, P.; Singh, E.M.; et al. The long non-coding RNA NEAT1 is responsive to neuronal activity and is associated with hyperexcitability states. Sci. Rep. 2017, 7, 40127. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Zhang, Y.; Lun, L.; Li, H.; Wang, Q.; Lin, J.; Tian, R.; Pan, H.; Zhang, H.; Chen, X. The Value of lncRNA NEAT1 as a Prognostic Factor for Survival of Cancer Outcome: A Meta-Analysis. Sci. Rep. 2017, 7, 13080. [Google Scholar] [CrossRef] [PubMed]
  46. Chen, T.; Wang, H.; Yang, P.; He, Z.Y. Prognostic role of long noncoding RNA NEAT1 in various carcinomas: A meta-analysis. Oncotargets Ther. 2017, 10, 993–1000. [Google Scholar] [CrossRef] [PubMed]
  47. Sun, S.J.; Lin, Q.; Ma, J.X.; Shi, W.W.; Yang, B.; Li, F. Long non-coding RNA NEAT1 acts as oncogene in NSCLC by regulating the Wnt signaling pathway. Eur. Rev. Med. Pharmacol. Sci. 2017, 21, 504–510. [Google Scholar]
  48. Jen, J.; Tang, Y.A.; Lu, Y.H.; Lin, C.C.; Lai, W.W.; Wang, Y.C. Oct4 transcriptionally regulates the expression of long non-coding RNAs NEAT1 and MALAT1 to promote lung cancer progression. Mol. Cancer 2017, 16, 104. [Google Scholar] [CrossRef]
  49. Zhang, M.; Wu, W.B.; Wang, Z.W.; Wang, X.H. lncRNA NEAT1 is closely related with progression of breast cancer via promoting proliferation and EMT. Eur. Rev. Med. Pharmacol. Sci. 2017, 21, 1020–1026. [Google Scholar]
  50. Wu, F.; Mo, Q.; Wan, X.; Dan, J.; Hu, H. NEAT1/has-mir-98-5p/MAPK6 axis is involved in non-small-cell lung cancer (NSCLC) development. J. Cell. Biochem. 2017. [Google Scholar] [CrossRef]
  51. Ke, H.; Zhao, L.; Feng, X.; Xu, H.; Zou, L.; Yang, Q.; Su, X.; Peng, L.; Jiao, B. NEAT1 is Required for Survival of Breast Cancer Cells Through FUS and miR-548. Gene Regul. Syst. Biol. 2016, 10, 11–17. [Google Scholar] [CrossRef] [PubMed]
  52. Lo, P.K.; Zhang, Y.; Wolfson, B.; Gernapudi, R.; Yao, Y.; Duru, N.; Zhou, Q. Dysregulation of the BRCA1/long non-coding RNA NEAT1 signaling axis contributes to breast tumorigenesis. Oncotarget 2016, 7, 65067–65089. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Li, W.; Zhang, Z.; Liu, X.; Cheng, X.; Zhang, Y.; Han, X.; Zhang, Y.; Liu, S.; Yang, J.; Xu, B.; et al. The FOXN3-NEAT1-SIN3A repressor complex promotes progression of hormonally responsive breast cancer. J. Clin. Investig. 2017, 127, 3421–3440. [Google Scholar] [CrossRef] [PubMed]
  54. Fang, L.; Sun, J.; Pan, Z.; Song, Y.; Zhong, L.; Zhang, Y.; Liu, Y.; Zheng, X.; Huang, P. Long non-coding RNA NEAT1 promotes hepatocellular carcinoma cell proliferation through the regulation of miR-129-5p-VCP-IkappaB. Am. J. Physiol. Gastrointest. Liver Physiol. 2017, 313, G150–G156. [Google Scholar] [CrossRef] [PubMed]
  55. Wang, Z.; Zou, Q.; Song, M.; Chen, J. NEAT1 promotes cell proliferation and invasion in hepatocellular carcinoma by negative regulating miR-613 expression. Biomed. Pharmacother. Biomed. Pharmacother. 2017, 94, 612–618. [Google Scholar] [CrossRef] [PubMed]
  56. Zhang, X.N.; Zhou, J.; Lu, X.J. The long noncoding RNA NEAT1 contributes to hepatocellular carcinoma development by sponging miR-485 and enhancing the expression of the STAT3. J. Cell. Physiol. 2018, 233, 6733–6741. [Google Scholar] [CrossRef]
  57. Tu, J.; Zhao, Z.; Xu, M.; Lu, X.; Chang, L.; Ji, J. NEAT1 upregulates TGF-beta1 to induce hepatocellular carcinoma progression by sponging hsa-mir-139-5p. J. Cell. Physiol. 2018. [Google Scholar] [CrossRef]
  58. Chen, Z.J.; Zhang, Z.; Xie, B.B.; Zhang, H.Y. Clinical significance of up-regulated lncRNA NEAT1 in prognosis of ovarian cancer. Eur. Rev. Med. Pharmacol. Sci. 2016, 20, 3373–3377. [Google Scholar]
  59. Ding, N.; Wu, H.; Tao, T.; Peng, E. NEAT1 regulates cell proliferation and apoptosis of ovarian cancer by miR-34a-5p/BCL2. Oncotargets Ther. 2017, 10, 4905–4915. [Google Scholar] [CrossRef]
  60. An, J.; Lv, W.; Zhang, Y. LncRNA NEAT1 contributes to paclitaxel resistance of ovarian cancer cells by regulating ZEB1 expression via miR-194. Oncotargets Ther. 2017, 10, 5377–5390. [Google Scholar] [CrossRef]
  61. Liu, Y.; Wang, Y.; Fu, X.; Lu, Z. Long non-coding RNA NEAT1 promoted ovarian cancer cells’ metastasis via regulating of miR-382-3p/ROCK1 axial. Cancer Sci. 2018. [Google Scholar] [CrossRef]
  62. Xiong, W.; Huang, C.; Deng, H.; Jian, C.; Zen, C.; Ye, K.; Zhong, Z.; Zhao, X.; Zhu, L. Oncogenic non-coding RNA NEAT1 promotes the prostate cancer cell growth through the SRC3/IGF1R/AKT pathway. Int. J. Biochem. Cell Biol. 2018, 94, 125–132. [Google Scholar] [CrossRef] [PubMed]
  63. Li, X.; Wang, X.; Song, W.; Xu, H.; Huang, R.; Wang, Y.; Zhao, W.; Xiao, Z.; Yang, X. Oncogenic properties of NEAT1 in prostate cancer cells depend on the CDC5L-AGRN transcriptional regulation circuit. Cancer Res. 2018. [Google Scholar] [CrossRef] [PubMed]
  64. Qi, X.; Zhang, D.H.; Wu, N.; Xiao, J.H.; Wang, X.; Ma, W. ceRNA in cancer: Possible functions and clinical implications. J. Med Genet. 2015, 52, 710–718. [Google Scholar] [CrossRef] [PubMed]
  65. Bartel, D.P. MicroRNAs: Target recognition and regulatory functions. Cell 2009, 136, 215–233. [Google Scholar] [CrossRef] [PubMed]
  66. Geng, H.; Tan, X.D. Functional diversity of long non-coding RNAs in immune regulation. Genes Dis. 2016, 3, 72–81. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Smolle, M.A.; Calin, H.N.; Pichler, M.; Calin, G.A. Noncoding RNAs and immune checkpoints-clinical implications as cancer therapeutics. FEBS J. 2017, 284, 1952–1966. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Hu, G.; Tang, Q.; Sharma, S.; Yu, F.; Escobar, T.M.; Muljo, S.A.; Zhu, J.; Zhao, K. Expression and regulation of intergenic long noncoding RNAs during T cell development and differentiation. Nat. Immunol. 2013, 14, 1190–1198. [Google Scholar] [CrossRef] [Green Version]
  69. Ranzani, V.; Rossetti, G.; Panzeri, I.; Arrigoni, A.; Bonnal, R.J.; Curti, S.; Gruarin, P.; Provasi, E.; Sugliano, E.; Marconi, M.; et al. The long intergenic noncoding RNA landscape of human lymphocytes highlights the regulation of T cell differentiation by linc-MAF-4. Nat. Immunol. 2015, 16, 318–325. [Google Scholar] [CrossRef] [Green Version]
  70. Venkatraman, A.; He, X.C.; Thorvaldsen, J.L.; Sugimura, R.; Perry, J.M.; Tao, F.; Zhao, M.; Christenson, M.K.; Sanchez, R.; Yu, J.Y.; et al. Maternal imprinting at the H19-Igf2 locus maintains adult haematopoietic stem cell quiescence. Nature 2013, 500, 345–349. [Google Scholar] [CrossRef]
  71. Wang, P.; Xue, Y.; Han, Y.; Lin, L.; Wu, C.; Xu, S.; Jiang, Z.; Xu, J.; Liu, Q.; Cao, X. The STAT3-binding long noncoding RNA lnc-DC controls human dendritic cell differentiation. Science 2014, 344, 310–313. [Google Scholar] [CrossRef] [PubMed]
  72. Kotzin, J.J.; Spencer, S.P.; McCright, S.J.; Kumar, D.B.U.; Collet, M.A.; Mowel, W.K.; Elliott, E.N.; Uyar, A.; Makiya, M.A.; Dunagin, M.C.; et al. The long non-coding RNA Morrbid regulates Bim and short-lived myeloid cell lifespan. Nature 2016, 537, 239–243. [Google Scholar] [CrossRef] [PubMed]
  73. Spurlock, C.F., 3rd; Tossberg, J.T.; Guo, Y.; Collier, S.P.; Crooke, P.S., 3rd; Aune, T.M. Expression and functions of long noncoding RNAs during human T helper cell differentiation. Nat. Commun. 2015, 6, 6932. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Carpenter, S.; Aiello, D.; Atianand, M.K.; Ricci, E.P.; Gandhi, P.; Hall, L.L.; Byron, M.; Monks, B.; Henry-Bezy, M.; Lawrence, J.B.; et al. A long noncoding RNA mediates both activation and repression of immune response genes. Science 2013, 341, 789–792. [Google Scholar] [CrossRef] [PubMed]
  75. Chen, Y.G.; Satpathy, A.T.; Chang, H.Y. Gene regulation in the immune system by long noncoding RNAs. Nat. Immunol. 2017, 18, 962–972. [Google Scholar] [CrossRef] [PubMed]
  76. Zeng, C.; Xu, Y.; Xu, L.; Yu, X.; Cheng, J.; Yang, L.; Chen, S.; Li, Y. Inhibition of long non-coding RNA NEAT1 impairs myeloid differentiation in acute promyelocytic leukemia cells. BMC Cancer 2014, 14, 693. [Google Scholar] [CrossRef]
  77. Giza, D.E.; Fuentes-Mattei, E.; Bullock, M.D.; Tudor, S.; Goblirsch, M.J.; Fabbri, M.; Lupu, F.; Yeung, S.J.; Vasilescu, C.; Calin, G.A. Cellular and viral microRNAs in sepsis: Mechanisms of action and clinical applications. Cell Death Differ. 2016, 23, 1906–1918. [Google Scholar] [CrossRef]
  78. Fuentes-Mattei, E.; Giza, D.E.; Shimizu, M.; Ivan, C.; Manning, J.T.; Tudor, S.; Ciccone, M.; Kargin, O.A.; Zhang, X.; Mur, P.; et al. Plasma Viral miRNAs Indicate a High Prevalence of Occult Viral Infections. EBioMedicine 2017, 20, 182–192. [Google Scholar] [CrossRef]
  79. Huang, S.; Qian, K.; Zhu, Y.; Huang, Z.; Luo, Q.; Qing, C. Diagnostic Value of the lncRNA NEAT1 in Peripheral Blood Mononuclear Cells of Patients with Sepsis. Dis. Markers 2017, 2017, 7962836. [Google Scholar] [CrossRef]
  80. Chen, Y.; Qiu, J.; Chen, B.; Lin, Y.; Chen, Y.; Xie, G.; Qiu, J.; Tong, H.; Jiang, D. Long non-coding RNA NEAT1 plays an important role in sepsis-induced acute kidney injury by targeting miR-204 and modulating the NF-kappaB pathway. Int. Immunopharmacol. 2018, 59, 252–260. [Google Scholar] [CrossRef]
  81. Li, X.Y.; Zhang, K.; Jiang, Z.Y.; Cai, L.H. MiR-204/miR-211 downregulation contributes to candidemia-induced kidney injuries via derepression of Hmx1 expression. Life Sci. 2014, 102, 139–144. [Google Scholar] [CrossRef] [PubMed]
  82. Abraham, E. Nuclear factor-kappaB and its role in sepsis-associated organ failure. J. Infect. Dis. 2003, 187 (Suppl. 2), S364–S369. [Google Scholar] [CrossRef]
  83. Zhang, Q.; Chen, C.Y.; Yedavalli, V.S.; Jeang, K.T. NEAT1 long noncoding RNA and paraspeckle bodies modulate HIV-1 posttranscriptional expression. mBio 2013, 4, e00596-12. [Google Scholar] [CrossRef] [PubMed]
  84. Budhiraja, S.; Liu, H.; Couturier, J.; Malovannaya, A.; Qin, J.; Lewis, D.E.; Rice, A.P. Mining the human complexome database identifies RBM14 as an XPO1-associated protein involved in HIV-1 Rev function. J. Virol. 2015, 89, 3557–3567. [Google Scholar] [CrossRef] [PubMed]
  85. Pandey, A.D.; Goswami, S.; Shukla, S.; Das, S.; Ghosal, S.; Pal, M.; Bandyopadhyay, B.; Ramachandran, V.; Basu, N.; Sood, V.; et al. Correlation of altered expression of a long non-coding RNA, NEAT1, in peripheral blood mononuclear cells with dengue disease progression. J. Infect. 2017, 75, 541–554. [Google Scholar] [CrossRef] [PubMed]
  86. Adriaens, C.; Standaert, L.; Barra, J.; Latil, M.; Verfaillie, A.; Kalev, P.; Boeckx, B.; Wijnhoven, P.W.; Radaelli, E.; Vermi, W.; et al. p53 induces formation of NEAT1 lncRNA-containing paraspeckles that modulate replication stress response and chemosensitivity. Nat. Med. 2016, 22, 861–868. [Google Scholar] [CrossRef] [PubMed]
  87. Yoneyama, M.; Kikuchi, M.; Natsukawa, T.; Shinobu, N.; Imaizumi, T.; Miyagishi, M.; Taira, K.; Akira, S.; Fujita, T. The RNA helicase RIG-I has an essential function in double-stranded RNA-induced innate antiviral responses. Nat. Immunol. 2004, 5, 730–737. [Google Scholar] [CrossRef] [PubMed]
  88. Loo, Y.M.; Gale, M., Jr. Immune signaling by RIG-I-like receptors. Immunity 2011, 34, 680–692. [Google Scholar] [CrossRef] [PubMed]
  89. Ma, H.; Han, P.; Ye, W.; Chen, H.; Zheng, X.; Cheng, L.; Zhang, L.; Yu, L.; Wu, X.; Xu, Z.; et al. The Long Noncoding RNA NEAT1 Exerts Antihantaviral Effects by Acting as Positive Feedback for RIG-I Signaling. J. Virol. 2017, 91, JVI-02250. [Google Scholar] [CrossRef] [PubMed]
  90. Beeharry, Y.; Goodrum, G.; Imperiale, C.J.; Pelchat, M. The Hepatitis Delta Virus accumulation requires paraspeckle components and affects NEAT1 level and PSP1 localization. Sci. Rep. 2018, 8, 6031. [Google Scholar] [CrossRef] [PubMed]
  91. Imamura, K.; Imamachi, N.; Akizuki, G.; Kumakura, M.; Kawaguchi, A.; Nagata, K.; Kato, A.; Kawaguchi, Y.; Sato, H.; Yoneda, M.; et al. Long noncoding RNA NEAT1-dependent SFPQ relocation from promoter region to paraspeckle mediates IL8 expression upon immune stimuli. Mol. Cell 2014, 53, 393–406. [Google Scholar] [CrossRef] [PubMed]
  92. Viollet, C.; Davis, D.A.; Tekeste, S.S.; Reczko, M.; Ziegelbauer, J.M.; Pezzella, F.; Ragoussis, J.; Yarchoan, R. RNA Sequencing Reveals that Kaposi Sarcoma-Associated Herpesvirus Infection Mimics Hypoxia Gene Expression Signature. PLoS Pathog. 2017, 13, e1006143. [Google Scholar] [CrossRef]
  93. Wang, Z.; Fan, P.; Zhao, Y.; Zhang, S.; Lu, J.; Xie, W.; Jiang, Y.; Lei, F.; Xu, N.; Zhang, Y. NEAT1 modulates herpes simplex virus-1 replication by regulating viral gene transcription. Cell. Mol. Life Sci. 2017, 74, 1117–1131. [Google Scholar] [CrossRef] [PubMed]
  94. Choudhry, H.; Albukhari, A.; Morotti, M.; Haider, S.; Moralli, D.; Smythies, J.; Schodel, J.; Green, C.M.; Camps, C.; Buffa, F.; et al. Tumor hypoxia induces nuclear paraspeckle formation through HIF-2alpha dependent transcriptional activation of NEAT1 leading to cancer cell survival. Oncogene 2015, 34, 4546. [Google Scholar] [CrossRef]
  95. Riva, P.; Ratti, A.; Venturin, M. The Long Non-Coding RNAs in Neurodegenerative Diseases: Novel Mechanisms of Pathogenesis. Curr. Alzheimer Res. 2016, 13, 1219–1231. [Google Scholar] [CrossRef]
  96. Li, D.; Yang, M.Q. Identification and characterization of conserved lncRNAs in human and rat brain. BMC Bioinform. 2017, 18, 489. [Google Scholar] [CrossRef]
  97. D’Haene, E.; Jacobs, E.Z.; Volders, P.J.; De Meyer, T.; Menten, B.; Vergult, S. Identification of long non-coding RNAs involved in neuronal development and intellectual disability. Sci. Rep. 2016, 6, 28396. [Google Scholar] [CrossRef] [PubMed]
  98. Ramos, A.D.; Diaz, A.; Nellore, A.; Delgado, R.N.; Park, K.Y.; Gonzales-Roybal, G.; Oldham, M.C.; Song, J.S.; Lim, D.A. Integration of genome-wide approaches identifies lncRNAs of adult neural stem cells and their progeny in vivo. Cell Stem Cell 2013, 12, 616–628. [Google Scholar] [CrossRef]
  99. Quan, Z.; Zheng, D.; Qing, H. Regulatory Roles of Long Non-Coding RNAs in the Central Nervous System and Associated Neurodegenerative Diseases. Front. Cell Neurosci. 2017, 11, 175. [Google Scholar] [CrossRef]
  100. Scheele, C.; Petrovic, N.; Faghihi, M.A.; Lassmann, T.; Fredriksson, K.; Rooyackers, O.; Wahlestedt, C.; Good, L.; Timmons, J.A. The human PINK1 locus is regulated in vivo by a non-coding natural antisense RNA during modulation of mitochondrial function. BMC Genom. 2007, 8, 74. [Google Scholar] [CrossRef] [PubMed]
  101. Chiba, M.; Kiyosawa, H.; Hiraiwa, N.; Ohkohchi, N.; Yasue, H. Existence of Pink1 antisense RNAs in mouse and their localization. Cytogenet. Genome Res. 2009, 126, 259–270. [Google Scholar] [CrossRef] [PubMed]
  102. Morais, V.A.; Verstreken, P.; Roethig, A.; Smet, J.; Snellinx, A.; Vanbrabant, M.; Haddad, D.; Frezza, C.; Mandemakers, W.; Vogt-Weisenhorn, D.; et al. Parkinson’s disease mutations in PINK1 result in decreased Complex I activity and deficient synaptic function. Embo Mol. Med. 2009, 1, 99–111. [Google Scholar] [CrossRef] [PubMed]
  103. Noble, E.E.; Billington, C.J.; Kotz, C.M.; Wang, C. The lighter side of BDNF. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2011, 300, R1053–R1069. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Xie, Y.; Hayden, M.R.; Xu, B. BDNF overexpression in the forebrain rescues Huntington’s disease phenotypes in YAC128 mice. J. Neurosci. 2010, 30, 14708–14718. [Google Scholar] [CrossRef]
  105. Wan, P.; Su, W.; Zhuo, Y. The Role of Long Noncoding RNAs in Neurodegenerative Diseases. Mol. Neurobiol. 2017, 54, 2012–2021. [Google Scholar] [CrossRef] [PubMed]
  106. Michelhaugh, S.K.; Lipovich, L.; Blythe, J.; Jia, H.; Kapatos, G.; Bannon, M.J. Mining Affymetrix microarray data for long non-coding RNAs: Altered expression in the nucleus accumbens of heroin abusers. J. Neurochem. 2011, 116, 459–466. [Google Scholar] [CrossRef] [PubMed]
  107. Zhong, J.; Jiang, L.; Huang, Z.; Zhang, H.; Cheng, C.; Liu, H.; He, J.; Wu, J.; Darwazeh, R.; Wu, Y.; et al. The long non-coding RNA Neat1 is an important mediator of the therapeutic effect of bexarotene on traumatic brain injury in mice. Brain Behav. Immun. 2017, 65, 183–194. [Google Scholar] [CrossRef] [PubMed]
  108. Bates, G.P. History of genetic disease: The molecular genetics of Huntington disease—A history. Nat. Rev. Genet. 2005, 6, 766–773. [Google Scholar] [CrossRef]
  109. Sunwoo, J.S.; Lee, S.T.; Im, W.; Lee, M.; Byun, J.I.; Jung, K.H.; Park, K.I.; Jung, K.Y.; Lee, S.K.; Chu, K.; et al. Altered Expression of the Long Noncoding RNA NEAT1 in Huntington’s Disease. Mol. Neurobiol. 2017, 54, 1577–1586. [Google Scholar] [CrossRef]
  110. Johnson, R. Long non-coding RNAs in Huntington’s disease neurodegeneration. Neurobiol. Dis. 2012, 46, 245–254. [Google Scholar] [CrossRef]
  111. Goldenberg, M.M. Multiple sclerosis review. P T 2012, 37, 175–184. [Google Scholar] [PubMed]
  112. Santoro, M.; Nociti, V.; Lucchini, M.; De Fino, C.; Losavio, F.A.; Mirabella, M. Expression Profile of Long Non-Coding RNAs in Serum of Patients with Multiple Sclerosis. J. Mol. Neurosci. 2016, 59, 18–23. [Google Scholar] [CrossRef] [PubMed]
  113. Lund, B.T.; Ashikian, N.; Ta, H.Q.; Chakryan, Y.; Manoukian, K.; Groshen, S.; Gilmore, W.; Cheema, G.S.; Stohl, W.; Burnett, M.E.; et al. Increased CXCL8 (IL-8) expression in Multiple Sclerosis. J. Neuroimmunol. 2004, 155, 161–171. [Google Scholar] [CrossRef] [PubMed]
  114. Bsibsi, M.; Bajramovic, J.J.; Vogt, M.H.; van Duijvenvoorden, E.; Baghat, A.; Persoon-Deen, C.; Tielen, F.; Verbeek, R.; Huitinga, I.; Ryffel, B.; et al. The microtubule regulator stathmin is an endogenous protein agonist for TLR3. J. Immunol. 2010, 184, 6929–6937. [Google Scholar] [CrossRef] [PubMed]
  115. Zarei, S.; Carr, K.; Reiley, L.; Diaz, K.; Guerra, O.; Altamirano, P.F.; Pagani, W.; Lodin, D.; Orozco, G.; Chinea, A. A comprehensive review of amyotrophic lateral sclerosis. Surg. Neurol. Int. 2015, 6, 171. [Google Scholar] [CrossRef] [PubMed]
  116. Nishimoto, Y.; Nakagawa, S.; Hirose, T.; Okano, H.J.; Takao, M.; Shibata, S.; Suyama, S.; Kuwako, K.; Imai, T.; Murayama, S.; et al. The long non-coding RNA nuclear-enriched abundant transcript 1_2 induces paraspeckle formation in the motor neuron during the early phase of amyotrophic lateral sclerosis. Mol. Brain 2013, 6, 31. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Kabashi, E.; Valdmanis, P.N.; Dion, P.; Spiegelman, D.; McConkey, B.J.; Vande Velde, C.; Bouchard, J.P.; Lacomblez, L.; Pochigaeva, K.; Salachas, F.; et al. TARDBP mutations in individuals with sporadic and familial amyotrophic lateral sclerosis. Nat. Genet. 2008, 40, 572–574. [Google Scholar] [CrossRef]
  118. Kwiatkowski, T.J., Jr.; Bosco, D.A.; Leclerc, A.L.; Tamrazian, E.; Vanderburg, C.R.; Russ, C.; Davis, A.; Gilchrist, J.; Kasarskis, E.J.; Munsat, T.; et al. Mutations in the FUS/TLS gene on chromosome 16 cause familial amyotrophic lateral sclerosis. Science 2009, 323, 1205–1208. [Google Scholar] [CrossRef]
  119. Chesselet, M.F. Dopamine and Parkinson’s disease: Is the killer in the house? Mol. Psychiatry 2003, 8, 369–370. [Google Scholar] [CrossRef]
  120. Pickrell, A.M.; Youle, R.J. The roles of PINK1, parkin, and mitochondrial fidelity in Parkinson’s disease. Neuron 2015, 85, 257–273. [Google Scholar] [CrossRef]
  121. Yan, W.; Chen, Z.Y.; Chen, J.Q.; Chen, H.M. LncRNA NEAT1 promotes autophagy in MPTP-induced Parkinson’s disease through stabilizing PINK1 protein. Biochem. Biophys. Res. Commun. 2018, 496, 1019–1024. [Google Scholar] [CrossRef] [PubMed]
  122. Liu, Y.; Lu, Z. Long non-coding RNA NEAT1 mediates the toxic of Parkinson’s disease induced by MPTP/MPP+ via regulation of gene expression. Clin. Exp. Pharmacol. Physiol. 2018. [Google Scholar] [CrossRef] [PubMed]
  123. Wang, X.; Xu, Y.; Zhu, Y.C.; Wang, Y.K.; Li, J.; Li, X.Y.; Ji, T.; Bai, S.J. LncRNA NEAT1 promotes extracellular matrix accumulation and epithelial-to-mesenchymal transition by targeting miR-27b-3p and ZEB1 in diabetic nephropathy. J. Cell. Physiol. 2018. [Google Scholar] [CrossRef] [PubMed]
  124. Wang, L.; Xia, J.W.; Ke, Z.P.; Zhang, B.H. Blockade of NEAT1 represses inflammation response and lipid uptake via modulating miR-342-3p in human macrophages THP-1 cells. J. Cell. Physiol. 2019, 234, 5319–5326. [Google Scholar] [CrossRef] [PubMed]
  125. Chen, D.D.; Hui, L.L.; Zhang, X.C.; Chang, Q. NEAT1 contributes to ox-LDL-induced inflammation and oxidative stress in macrophages through inhibiting miR-128. J. Cell. Biochem. 2018. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Nuclear enriched abundant transcript 1 (NEAT1)’s domain architecture and schematic paraspeckle structure. (A) The long isoform of NEAT1 (NEAT1_2) contains three domains which are relevant for binding the paraspeckle-associated proteins 54 kDa nuclear RNA- and DNA-binding protein/non-POU domain-containing octamer-binding protein (p54nrb/NONO; orange), paraspeckle component 1 (PSPC1; yellow), and polypyrimidine tract-binding protein PTB-associated splicing factor/splicing factor proline glutamine rich (PSF/SFPQ; brown). p54nrb/NONO and PSF/SFPQ directly interact with NEAT1’s middle domain, whereas three protein interaction sites near the 5’ and the 3’ end facilitate binding of p54nrb/NONO. All three abovementioned proteins form heterodimers in every possible combination, and thus contribute to the formation of paraspeckles. Only proven interactions of proteins with NEAT1 and each other are shown. (B) Paraspeckles are arranged in a spheroidal, highly ordered structure with NEAT1’s middle domain being located in the center while its 5’ and 3’ termini are at the periphery of the structure. Paraspeckle-associated proteins p54nrb/NONO, PSF/SFPQ, and PSPC1 (as well as other paraspeckle proteins; not shown in figure) are distributed within the structure in consideration of the before established binding domains on NEAT1.
Figure 1. Nuclear enriched abundant transcript 1 (NEAT1)’s domain architecture and schematic paraspeckle structure. (A) The long isoform of NEAT1 (NEAT1_2) contains three domains which are relevant for binding the paraspeckle-associated proteins 54 kDa nuclear RNA- and DNA-binding protein/non-POU domain-containing octamer-binding protein (p54nrb/NONO; orange), paraspeckle component 1 (PSPC1; yellow), and polypyrimidine tract-binding protein PTB-associated splicing factor/splicing factor proline glutamine rich (PSF/SFPQ; brown). p54nrb/NONO and PSF/SFPQ directly interact with NEAT1’s middle domain, whereas three protein interaction sites near the 5’ and the 3’ end facilitate binding of p54nrb/NONO. All three abovementioned proteins form heterodimers in every possible combination, and thus contribute to the formation of paraspeckles. Only proven interactions of proteins with NEAT1 and each other are shown. (B) Paraspeckles are arranged in a spheroidal, highly ordered structure with NEAT1’s middle domain being located in the center while its 5’ and 3’ termini are at the periphery of the structure. Paraspeckle-associated proteins p54nrb/NONO, PSF/SFPQ, and PSPC1 (as well as other paraspeckle proteins; not shown in figure) are distributed within the structure in consideration of the before established binding domains on NEAT1.
Ijms 20 00627 g001
Figure 2. Role of NEAT1 in the regulation of gene expression. NEAT1 influences gene regulation through two predominant functions. On one hand, NEAT1-dependent paraspeckle formation leads to the sequestration of paraspeckle proteins such as PSPC1, PSF/SFPQ, and p54nrb/NONO, therefore limiting their effect on the transcriptional regulation. On the other hand, adenosine-to-inosine (A–I) edited mRNAs are more efficiently bound by formed paraspeckles and in turn more effectively retained in the nucleus, instead of being transported to the cytoplasm.
Figure 2. Role of NEAT1 in the regulation of gene expression. NEAT1 influences gene regulation through two predominant functions. On one hand, NEAT1-dependent paraspeckle formation leads to the sequestration of paraspeckle proteins such as PSPC1, PSF/SFPQ, and p54nrb/NONO, therefore limiting their effect on the transcriptional regulation. On the other hand, adenosine-to-inosine (A–I) edited mRNAs are more efficiently bound by formed paraspeckles and in turn more effectively retained in the nucleus, instead of being transported to the cytoplasm.
Ijms 20 00627 g002
Table 1. NEAT1 expression levels in several viral-induced diseases together with the proposed molecular pathway.
Table 1. NEAT1 expression levels in several viral-induced diseases together with the proposed molecular pathway.
NEAT1 ExpressionViral DiseasePro-viral or Anti-viralPathwayLiterature
UpregulationHIV-1anti-viralRev-dependent nuclear export[83,84]
Mild dengue [85]
Herpes simplexpro-viralP54nrb, PSPC1[91,93]
Hantavirusanti-viralRIG-I-signaling[88,89]
Hepatitis Danti-viralIL-8 induction[90]
Influenzaanti-viralIL-8 induction by SFPQ inhibition[91]
Down-regulationSevere dengueanti-viralp53 induced apoptosis[85]
Table 2. NEAT1 expression levels in several neurodegenerative diseases and predicted co-players.
Table 2. NEAT1 expression levels in several neurodegenerative diseases and predicted co-players.
NEAT1 ExpressionNeurodegenerative DiseaseCo-playersLiterature
UpregulationHuntington’s DiseaseREST, p53[109,110]
Multiple SclerosisIL-8, stathmin & TLR3[112,113,114]
Amyotrophic Lateral SclerosisTDP-43, FUS/TLS[116]
Parkinson’s Diseaseα-synuclein[121,122]

Share and Cite

MDPI and ACS Style

Prinz, F.; Kapeller, A.; Pichler, M.; Klec, C. The Implications of the Long Non-Coding RNA NEAT1 in Non-Cancerous Diseases. Int. J. Mol. Sci. 2019, 20, 627. https://doi.org/10.3390/ijms20030627

AMA Style

Prinz F, Kapeller A, Pichler M, Klec C. The Implications of the Long Non-Coding RNA NEAT1 in Non-Cancerous Diseases. International Journal of Molecular Sciences. 2019; 20(3):627. https://doi.org/10.3390/ijms20030627

Chicago/Turabian Style

Prinz, Felix, Anita Kapeller, Martin Pichler, and Christiane Klec. 2019. "The Implications of the Long Non-Coding RNA NEAT1 in Non-Cancerous Diseases" International Journal of Molecular Sciences 20, no. 3: 627. https://doi.org/10.3390/ijms20030627

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop