Abstract
Long non-coding RNAs (lncRNAs) are potent regulators of immune cell development and function. Their implication in multiple immune-mediated disorders highlights lncRNAs as exciting biomarkers and potential drug targets. Recent technological innovations in oligo-based therapeutics, development of RNA-targeting small molecules, and CRISPR-based approaches, position RNA as the next therapeutic frontier. Here, we review the latest advances made toward understanding the role of lncRNAs in human immunological disorders and further discuss RNA-targeting approaches that could be potentially exploited to manipulate lncRNA function as a clinical intervention.
Introduction
Although the human genome is pervasively transcribed into RNA, less than 2% of our DNA encodes proteins through messenger RNAs (mRNAs)[1,2]. Moreover, non-coding RNAs (ncRNAs) account for up to 97% of the mass of RNA in the cell, with rRNAs and tRNAs being the most abundant transcripts[3]. These ncRNAs show a rich diversity in both form and function, and based on their size, they are broadly classified into small non-coding RNAs (≤ 200 nucleotides reviewed in[4-7]) and long non-coding RNAs (lncRNAs; > 200 nucleotides) [8]. The varied molecular functions of lncRNAs include—but are not limited to—the regulation of transcription, transcript stability, translation, and cell signaling, as well as nuclear scaffolding. Moreover, the NONCODE database currently boasts 96,000 human lncRNA genes, indicating that lncRNAs potentially outnumber protein-coding (PC) genes by as many as five-fold[9]. Although, by definition, lncRNAs do not encode proteins, many have molecular features similar to that of PC mRNAs in that they are spliced, 3’ polyadenylated, and 5’ m7G capped. However, it should be noted that a small fraction of transcripts previously annotated as lncRNAs contain small open reading frames that could be translated into small peptides or micropeptides, some of which have biological function[10-12]. Conversely, some PC genes produce transcripts that act similar to ncRNAs, blurring the line between coding and noncoding genes[13-15]. While the functions of most lncRNAs are largely unknown, they play key roles in virtually all major subclasses of immune cells, such as coordinating immune cell differentiation, homeostasis, and effector function[16-28]. (Recent reviews elaborate on the mechanisms of lncRNAs implicated in the biology of immune cells[29-32].) Compared to mRNAs, lncRNAs are generally expressed in a more tissue-, cell type-, and disease-specific manner[33-35], making them attractive therapeutic targets and biomarkers for disease diagnosis. For example, overexpression of the lncRNA PCA3 in prostate cancer cells is currently exploited as a urine-accessible biomarker in the differential diagnosis of prostate cancer[36,37]. Thus, characterizing lncRNAs in immune cells will not only aid in understanding the function of the non-coding portions of the human genome, but may identify biomarkers for immunologic diseases and eventually reveal novel “druggable” targets.
lncRNAs in immunologic diseases
Transcriptome analyses of various immune cells have produced a catalog of lncRNAs that are differentially expressed in a broad range of immune-related diseases. However, in most cases, the function of the lncRNAs are unknown and their contribution to disease pathogenesis have not been tested. In this review, we will describe a selection of the best-characterized immunologic disease-associated lncRNAs (additional examples are listed in Table 1).
Table 1: A selection of lncRNAs associated with various immune disorders.
The lncRNAs reported in this table have been curated based on their association with an immune related disorder, and have undergone additional characterization either in vivo or in vitro. Due to space restraints, the many disease associated lncRNAs that lack functional testing are not listed.
| LncRNA | Species Studied | Relevant cell(s) | Disease | Ref |
|---|---|---|---|---|
| Plasmacytoma variant translocation 1 (PVT1) | Human | Primary airway smooth muscle cells | Asthma | [112] |
| LNC_000127 | Human | PBMCs - Eosinophils | Asthma | [113] |
| BANCR | Human | Primary esophageal epithelial cells | Eosinophilic esophagitis | [114] |
| GAS5 | Mouse/Human | PBMCs | Rheumatoid arthritis (RA); systemic lupus erythematosus(SLE); multiple sclerosis(MS); sarcoidosis; Inflammatory bowel diseases (IDB) | [115,116] |
| HOTAIR | Human | PBMC | RA | [117] |
| H19 | Human | Isolated synovial macrophages (in RA) and intestinal epithelial cells (in IBD) | RA; IBD | [118,119] |
| NEAT1 | Human | Monocytes | SLE | [120] |
| Linc0949, and linc0597 | Human | PMBC | SLE | [121] |
| Psoriasis-associated non-protein coding RNA induced by stress (PRINS) | Human | Epidermal keratinocytes | Psoriasis | [122-124] |
| PSORS1C3 | Human | Tonsils and skin biopsies | Psoriasis | [125] |
| IFNG-AS1 | Human | CD4 T cells | IBD | [126] |
| CDKN2B-AS1 | Human | Intestinal epithelial cells | IBD | [127] |
| LINC00305 | Human | Monocytes | Atherosclerosis | [57] |
| TMEVPG1 aka NeST aka IFNG-AS1 | Human | Th1 cells | Hashimoto’s Thyroiditis | [18,28,128,129] |
| FlicR | Mouse/Human | Tregs | Diabetes | [130] |
| Linc-MAF-4 | Mouse/Human | Th1 | MS | [30,46] |
| Morrbid | Mouse/Human | Eosinophils, CD8+ T cells, monocytes | Hypereosinophilic Syndrome; Lymphocytic choriomeningitis virus infection; AML | [42,45,105] |
| Lnc13 | Human | Macrophages | CeD | [41] |
| LncRNA-CD244 | Human | CD8+ T cells | MTB infection | |
| NKILA | Human | Tumor infiltrating T cells | Cancer | [51] |
| THRIL | Human | Macrophages | Kawasaki disease | [26] |
Lnc13 in celiac disease
Celiac disease (CeD) is a complex, immune cell-mediated, gluten-sensitive enteropathy with varying clinical manifestations[38,39]. CeD has a strong genetic influence, and while risk variants in the major histocompatibility (MHC) region explain most CeD heritability, single-nucleotide polymorphisms (SNPs) outside of MHC are also implicated. Like other complex diseases, the majority of CeD-associated SNPs fall within non-coding regions[40]. While most complex disease SNPs likely alter the function of DNA regulatory elements, some may impact the function of lncRNAs. Indeed Castellanos-Rubio et al. discovered a novel lncRNA, called lnc13, that is expressed from a CeD-associated haplotype block[41]. Under homeostatic conditions in macrophages, lnc13 repressed expression of inflammatory genes by binding to their transcriptional start sites and recruiting the heterogeneous nuclear ribonucleoprotein hnRNPD. Intestinal biopsies from patients with CeD showed reduced levels of lnc13 RNA, which could potentially render these cells hyperinflammatory. Perhaps most interesting was the finding that lnc13 bound less efficiently to hnRNPD when it contained a CeD-associated SNP, thereby reducing its ability to suppress the inflammatory response. This study provides a lncRNA-based mechanism by which SNPs can contribute to CeD. Moreover, lnc13 serves as a clear example of how genetic changes can increase disease risk by directly impairing lncRNA function.
MORRBID in hypereosinophilic syndrome
The lncRNA Morrbid was first identified in mice as a regulator of short-lived myeloid cell lifespan[42]. In neutrophils, eosinophils, and monocytes, Morrbid represses expression of the pro-apoptotic gene Bcl2l11 (a.k.a Bim), which lies on the same chromosome approximately 150 kilobases away. Kotzin et al. found that Morrbid recruits the EZH2-containing polycomb repressive complex 2 (PRC2) to the Bcl2l11 promoter where it catalyzes deposition of H3K27me3 to maintain Bcl2l11 in a transcriptionally poised state[42]. The observation that Morrbid expression was induced by common β chain cytokines (IL-3, IL-5, and GM-CSF) suggests that Morrbid connects the presence of these external survival factors with the transcriptional program controlling cell lifespan. Morrbid is conserved across multiple species, and similar to mice, human MORRBID is most highly expressed in granulocytes. Moreover, MORRBID is overexpressed in eosinophils from individuals with hypereosinophilic syndrome, highlighting the relevance of MORRBID to human disease[42]. Although likely downstream of the elevated IL-5 levels that are observed in hypereosinophilic syndrome, the increased expression of MORRBID could potentially be increasing the steady-state number of eosinophils in these individuals by extending eosinophil lifespan, thereby promoting hypereosinophilia. If so, pharmacologic inhibition of MORRBID could potentially reduce eosinophil numbers. Indeed, clinical trials blocking IL-5 signaling have shown some efficacy in the treatment of hypereosinophilic syndrome[43,44]. A recent study of how inflammatory stress impacts hematopoiesis in mice has shown that Morrbid is also important in promoting the survival of pre-leukemic hematopoietic stem and progenitor cells (HSPCs)[45]. Interestingly, MORRBID overexpression is observed in a subset of acute myeloid leukemia patients and is associated with poor overall survival (R. Kapur and Z. Cai personal communication).
Linc-MAF-4 in multiple sclerosis
Linc-MAF-4 is a chromatin-associated lncRNA that is transcribed from a locus 139.5 kilobases upstream of the gene MAI, which encodes the C-Maf transcription factor that regulates Th2 cell differentiation[30]. Expression of these two genes is inversely correlated: linc-MAF-4 is expressed in Th1 cells whereas MAF is expressed in Th2 cells. In Th1 cells, Ranzani et al. found that linc-MAF-4 recruits EZH2 and LSD1 to the MAF promoter, leading to deposition of H3K27me3 and silencing of MAF[30]. Knockdown of linc-MAF-4 in Th1 cells resulted in inappropriate expression of MAF, GATA3, and IL4, suggesting that linc-MAF-4 promotes Th1 differentiation by blocking the C-Maf-controlled Th2 transcriptional program. A subsequent study connected linc-MAF-4 to multiple sclerosis (MS)[46]. MS is a disease of the central nervous system in which Th1 and Th17 cells play a critical role. Peripheral blood mononuclear cells (PBMCs) from MS patients exhibit increased expression of linc-MAF-4 and decreased expression of MAF, as compared to healthy controls[46]. Interestingly, higher expression levels of linc-MAF-4 positively correlate with an increased annual relapse rate, indicating that this lncRNA could potentially serve as a biomarker for MS. However, the cause for linc-MAF-4 upregulation in MS has not been established; therefore, determining whether elevated levels of linc-MAF-4 reflect increased numbers of Th1 cells or, alternatively, whether dysregulation of linc-MAF-4 could potentially contribute to MS susceptibility by promoting Th1 differentiation, is an area for further research.
LncRNA-CD244 in Mycobacterium tuberculosis infection
CD8+ T cells mount an adaptive immune response during infection with Mycobacterium tuberculosis (MTB)[47]. In humans, CD244 is a marker of CD8+ T cell exhaustion in chronic infection, and when highly expressed, it acts as an inhibitory receptor [48]. Wang et al. discovered lncRNA-CD244 as a negative regulator of anti-MTB responses[49]. Patients with an active MTB infection were found to have increased numbers of CD244+CD8+ T cells, and ex vivo stimulation of these cells with MTB lysates further increased CD244 expression. Blockade of CD244 signaling enhanced production of IFN-γ, TNF-α, and IL-6 by CD8+ T cells, confirming that CD244 upregulation impairs anti-MTB responses. In parallel, lncRNA-CD244 was found to be highly expressed in CD244+CD8+ T cells during active MTB infection, and expression of lncRNA-CD244 was induced by CD244 signaling. Wang et al. further demonstrated that lncRNA-CD244 bound EZH2 and mediated its recruitment to the IFNG and TNFA loci, inducing a repressive chromatin state. This finding provides a mechanism to explain how CD244 signaling suppresses CD8+ T cell responses. Finally, they showed that in a humanized mouse model, knockdown of lncRNA-CD244 in adoptively transferred CD8+ T cells significantly enhanced IFN-γ and TNF-α expression and improved protective immunity. Given that CD244 expression is implicated in impaired adaptive immune responses in chronic viral infections and cancer[48], lncRNA-CD244 could be a potential therapeutic target to limit CD8+ T cell exhaustion.
NKILA in tumor-infiltrating T cells
Discovered in a microarray screen of breast cancer cells treated with inflammatory cytokines, NF-κB interacting lncRNA (NKILA) was found to bind NF-κB/IκB and mask the phosphorylation motifs of IκB, thereby inhibiting NF-κB activation[50]. Low NKILA expression is associated with breast cancer metastasis and poor patient prognosis[51], and a similar association is observed in other cancers[52-55]. Recently, Huang et al. reported a separate role for NKILA in tumor-infiltrating cytotoxic T lymphocytes (CTLs) and type 1 helper T (Th1) cells[51]. They observed that subsets of tumor-infiltrating T cells displayed increased sensitivity to activation-induced cell death (AICD) that was in part due to NF-κB inactivation driven by upregulation of NKILA. Stimulating AICD is one of the many mechanisms tumors can use to evade the immune system[56], and this is the first report connecting this mechanism to a lncRNA. Importantly, Huang et al. found that patients with a greater percentage of tumor-infiltrating CTLs that expressed high levels of NKILA had shorter overall survival and disease-free survival than patients with tumor-infiltrating CTLs that expressed lower levels of NKILA. Using patient-derived breast cancer xenografts in mice, Huang et al. showed that knockdown of NKILA expression in CTLs increased their tumor infiltration, reduced AICD, and concomitantly inhibited cancer growth. These findings highlight the complex roles lncRNAs may play in disease, with increased expression of NKILA in immune cells and cancer cells being associated with opposing clinical outcomes, and moreover, suggest that therapeutic targeting of lncRNAs may necessitate a highly nuanced approach.
LINC00305 in atherosclerosis
By searching GWAS databases. Zhang et al. discovered atherosclerosis-associated SNPs located in the intron of the novel lncRNA LINC00305[57]. Interestingly, they showed that LINC00305 was expressed in monocytes and atherosclerotic plaques, and was overexpressed in PBMCs of individuals with atherosclerosis. Using a cDNA overexpression strategy in the monocytic cell line THP-1, they found that LINC00305 activated inflammation-associated genes through the NF-κB pathway. This suggests that LINC00305 is an immune lncRNA that promotes the inflammation underlying atherosclerosis. The authors next demonstrated that LINC00305 binds the lipocalin-interacting membrane receptor (LIMR) and increases its interaction with the aryl hydrocarbon receptor repressor (AHRR), which in turn is responsible for the augmented NF-κB activation. What remains to be addressed is how these intronic SNPs impact expression of LINC00305, and why this is specifically associated with atherosclerosis.
LncRNAs as therapeutic targets
Given the vast number of lncRNAs and their contribution to disease, development of lncRNA targeting strategies could greatly broaden the landscape of therapeutic interventions. While little has been done with respect to lncRNAs specifically, a number of approaches have been successfully developed to target mRNAs and microRNAs. In general, these approaches can drive RNA degradation, alter splicing patterns, or prevent translation of the target RNA. While lncRNAs do not undergo translation, these approaches may be adapted to prevent lncRNAs from binding to the protein(s) they interact with. Although some lncRNAs can be found in the cytoplasm, many are restricted to the nucleus[58]; therefore, care must be taken to select a targeting approach that is compatible with the subcellular localization of the target lncRNA (see below). Since most in vivo RNA-targeting modalities are currently delivered systemically (Figure 1), this provides a significant challenge in promoting efficient uptake by relevant cell types[59]. However, a unique property of the immune system can be exploited to partially circumvent these restrictions: for example, patient-derived HSPCs can be isolated and genetically modified ex vivo, expanded in number, and returned to the patient (Figure 1). Below, we present examples of different RNA-targeting strategies, as well as highlight some that are already in clinical use.
Figure 1. Potential strategies for therapeutic targeting of immune cell lncRNAs.
For transient targeting, ASOs are delivered systematically in vivo via intravenous or subcutaneous injections, or delivered to specific sites such as the cerebrospinal fluid. Following uptake by relevant cells types, ASOs alter lncRNA function by driving lncRNA degradation, altering splicing or blocking interaction with binding proteins. For long-term targeting, ex vivo editing of patient derived cells will likely be more efficacious. For example, hematopoietic stem cells can be harvested and edited ex vivo to manipulate expression of target lncRNAs, before delivery of corrected cells back to the patient. CRISPR based technologies are particularly amenable to this approach, with CRISPRi being used to silence lncRNA transcription or CasRx to knockdown the lncRNA transcripts. However, as both CRISPRi and CasRx require continued expression or CRISPR proteins special care must be taken to avoid toxicity or off-target effects.
RNA interference (RNAi)
Small interfering RNAs (siRNAs) are 21–23-nucleotide, double-stranded RNA molecules in which one strand is complementary to the target RNA. The antisense strand hybridizes to the target endogenous RNA in the cytoplasm and invokes a degradation pathway that involves dicer, RNA-induced silencing complex (RISC), and the AGO2 endonuclease[60]. Unmodified siRNAs are themselves susceptible to nucleases and have short half lives in vivo. The addition of 2′-O methyl sugar residues and phosphorothioate linkages to the 3′ end of the siRNA can slow enzymatic degradation, thereby increasing pharmacologic potential[61]. To increase the efficiency of delivery and uptake by relevant cell types, siRNAs can be delivered intravenously or intramuscularly with one of three different formulations: lipid nanoparticles[62], GalNAc conjugates[63], or masked endosomolytic compounds[64]. In 2018, the first siRNA drug, Onpattro™, was approved by the FDA to treat hereditary transthyretin amyloidosis[65]. Onpattro™ is formulated with nanoparticles and administered intravenously. Although not yet tested in humans, siRNA-mediated knockdown of the lncRNAs MALAT1 and HOTAIR in animal models has been shown to reduce cancer cell proliferation, growth, and metastasis, suggesting that siRNAs may also be used for lncRNA targeting [66,67].
Antisense oligos (ASOs)
ASOs are short 15–20-nucleotide, single-stranded DNA molecules that hybridize to endogenous target RNAs to initiate a ribonuclease (RNase) H-dependent degradation pathway[61,68,69]. RNA:DNA hybrids are substrates for RNase H1, which is concentrated inside the nucleus[70,71]. Unmodified, single-stranded ASOs are easily degraded, have poor bioavailability, and display suboptimal target binding. These shortcomings have been addressed by multiple advances in the chemical modification of ASOs, as reviewed in Shen et al. [72]. One prime example of a chemically-modified ASO is Spinraza™, an FDA-approved drug used in the treatment of spinal muscular atrophy (SMA). Spinraza™ is a phosphorothioate-linked, 2′-O–2-methoxyethyl-modified, ASO drug[73] that promotes alternative splicing of SMN2 mRNAs by targeting an intron splice silencer, thereby boosting production of the encoded full-length survival motor neuron protein[74]. One of the biggest challenges with developing efficacious ASO-based therapeutics is delivering them to target tissues and cell types. When given systemically, ASOs accumulate in the liver and kidneys [75-78]. Delivery of ASOs directly into target tissues can significantly increase uptake by relevant cell type[79]. Indeed, Spinraza™ must be administered directly into the cerebrospinal fluid. In addition, conjugation of ASOs to different biologic carriers has the potential to promote their uptake by cells of interest [79-82]. Finally, given ASOs ability to target nuclear RNAs, they are preferable to siRNAs for targeting most lncRNAs[70,71]. Indeed, the feasibility of using ASOs for in vivo lncRNA targeting has been demonstrated in different mouse models of cancer[83,84] and immune disorders[85-88].
(i). Gapmer, second generation ASOs
Gapmers are ASOs that contain a core DNA sequence flanked 5’ and 3’ by short RNA sequences consisting of chemically modified, degradation-resistant ribonucleotides. This RNA–DNA–RNA chimera enhances hybridization affinity to target RNA sequences and simultaneously serves as a substrate for RNase H1[89,90]. TEGSEDI™ is a 20-nucleotide, phosphorothioate-linked gapmer that has five 2′-O-–2-methoxyethy ribonucleotides at each terminus flanking a core of ten 2′-deoxyribonucleotides[91]. TEGSEDI™ was recently FDA-approved for the treatment of hereditary transthyretin amyloidosis[92]. In contrast to the siRNA Onpattro™, TEGSEDI™ is water-soluble and is administered subcutaneously without nanoparticle formulation[93,94]. No special delivery mechanisms are required for TEGSEDI™ as the target tissue is the liver.
(ii). Phosphorodiamidate morpholino oligomers and peptide nucleic acid, third generation ASOs
Phosphorodiamidate morpholino oligomers (PMOs) are charge-neutral, 20–25 nucleotide, single-stranded DNA analogs, where the five-membered ribofuranosyl rings of DNA are replaced with six-membered morpholino rings connected by phosphorodiamidate linkages[95]. PMOs effectively bind to target mRNAs to block translation through steric hindrance and thus work independently of RNase H-dependent degradation[96]. PMO-based EXONDYS 51™ is an FDA-approved therapy for Duchenne muscular dystrophy that functions by altering splicing of the dystrophin gene[97].
Peptide nucleic acids (PNAs), like PMOs, are charge-neutral oligomers with an N-(2-aminoethyl) glycine backbone. The neutral amide backbone makes PNAs resistant to both nucleases and proteases; thus PNA–DNA or PNA–RNA duplexes are more stable than the natural homo- or heteroduplexes[98]. Özcş et al. used PNAs to disrupt interactions of the lncRNA HOTAIR with PRC2, thereby reducing tumor progression and improving survival of mice harboring platinum-resistant, patient-derived ovarian tumor xenografts[99]. These initial studies demonstrate the feasibility of PNA-mediated targeting of lncRNAs in vivo.
Small molecules
These represent a diverse class of low-molecular-weight organic compounds that, by virtue of their small size and chemical properties, may easily enter cells where they can interact with and affect host molecules, such as proteins and RNAs. For example, ABX464 is small-molecule inhibitor that has shown efficacy in a phase 2a proof-of-concept study to treat ulcerative colitis. ABX464 binds the cap-binding complex (CBC) to trigger splicing of a lncRNA that houses the anti-inflammatory miR-124, thereby increasing miR-124 expression. In addition, several groups have developed small molecules against the lncRNAs BDNF-AS, HOTAIR, GAS5 and MALAT1, highlighting the lncRNA-targeting potential for this class of molecules[100-102]. While the development of RNA-targeting small molecules is relatively nascent, we predict that this field will flourish in the coming years and lead to the development of therapeutics against currently ‘undruggable’ targets.
CRISPR–Cas systems
CRISPR–Cas9 genome-editing technology depends on the ability to recruit the Cas9 bacterial endonuclease to specific genomic sites via the sequence-complementarity of its associated guide RNAs (gRNA). The gRNAs are designed to the target DNA sequences and bind downstream of a protospacer adjacent motif (PAM; three-nucleotide sequence). Cas9 then recognizes the PAM sequence and generates a double-stranded DNA break three nucleotides upstream[103,104]. Non-homology-directed repair of the double-stranded break by the cell’s intrinsic DNA repair machinery leaves behind mutations in the targeted DNA. While such mutations can effectively silence PC genes by inducing frameshifts in the coding sequence, the impact of non-homology-directed repair on lncRNAs is less predictable. To circumvent this, we and others have used CRISPR–Cas9 to delete either lncRNA promoter sequences or entire lncRNA loci to prevent their expression[105,106]. However, this approach is not without caveats. For example, lncRNA loci could contain DNA regulatory elements that regulate local gene expression. Furthermore, there is substantial crosstalk between promoters of different genes [107-109]. Therefore, for each genomic deletion, it is critical to assess the impact on local gene expression to account for unwanted off-target effects. Alternatively, CRISPR–Cas9-based epigenome-editing methods can be used to modify expression of lncRNAs using CRISPR interference/activation (CRISPRi/a). Using a catalytically inactive Cas9 that is fused to either a transcriptional repressor (KRAB domain) or activators (VP64/P300 domains), a gene promoter can be targeted using gRNAs to achieve specific transcriptional interference or activation[110]. Recently, Konermann et al. identified a Type VI-D CRISPR–Cas system, called CasRx, that allows direct targeting of transcripts[111]. They demonstrated that CasRx was able to efficiently knockdown both mRNA and lncRNA targets[111]. While, both CRISPRi/a and CasRx are still in their infancy, they provide an exciting area to be explored for therapeutic targeting of lncRNAs. As of November 2019, there are five active CRISPR–Cas9 clinical trials in the US, and four on-going and one completed trial in China; of these 10, nine trials are hematopoietic cell-specific. Phase-I/II clinical trials are being actively pursued to treat severe sickle cell disease ( NCT03745287) and transfusion-dependent β-thalassemia ( NCT03655678) by using CRISPR-Cas9 to target a BCL11A gene enhancer in HSPCs. Interestingly, seven Phase 1/2 clinical trials are underway using CRISPR-Cas9-engineered T Cells, notably for treating B-cell malignancies ( NCT03044743, NCT04035434, and NCT03398967), mesothelin-positive solid tumors ( NCT03747965 and NCT03545815), leukemia and lymphoma ( NCT03166878 and NCT03044743), and carcinomas ( NCT03044743 and NCT03081715). These clinical trials highlight the feasibility of applying CRISPR–Cas technology to hematopoietic cells as a therapeutic tool.
Conclusion
The lncRNA field has expanded rapidly over the last decade with lncRNAs emerging as important molecules in immune regulation and diverse diseases. However, for lncRNA therapeutics to be realized, we must first identify those that truly drive pathogenesis. Furthermore, although lncRNAs are often expressed in a cell type-specific manner, some—such as in the case of NKILA—may play opposing roles in different cell types even within a single disease. The biggest challenge will therefore be to develop technologies that will enable delivery of RNA targeting therapeutics in a cell type-selective manner.
Highlights.
More than 97% of transcripts in the cell are categorized as non-coding RNAs.
long non-coding RNAs (lncRNAs) outnumber protein coding genes.
lncRNAs are often expressed in a tissue-, cell type-, and disease-specific manner.
lncRNAs play known roles in virtually all major subclasses of immune cells.
lncRNAs are implicated in multiple immunologic disorders.
Acknowledgements
AW was supported by NIH R01AI141609, R21AI13522, and R21AI133440. We would like to thank Carmen Robinett for editing the manuscript, and Matt Wimsatt for illustrations.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- [1].Pertea M, The human transcriptome: An unfinished story, Genes (Basel). (2012). doi: 10.3390/genes3030344. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [2].Hangauer MJ, Vaughn IW, McManus MT, Pervasive Transcription of the Human Genome Produces Thousands of Previously Unidentified Long Intergenic Noncoding RNAs, PLoS Genet. (2013). doi: 10.1371/journal.pgen.1003569. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [3].Palazzo AF, Uee ES, Non-coding RNA: What is functional and what is junk?, Front. Genet (2015). doi: 10.3389/fgene.2015.00002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [4].Carthew RW, Sontheimer EJ, Origins and Mechanisms of miRNAs and siRNAs, Cell. (2009). doi: 10.1016/j.cell.2009.01.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [5].Treiber T, Treiber N, Meister G, Regulation of microRNA biogenesis and its crosstalk with other cellular pathways, Nat. Rev. Mol. Cell Biol (2019). doi: 10.1038/s41580-018-0059-1. [DOI] [PubMed] [Google Scholar]
- [6].Iwasaki YW, Siomi MC, Siomi H, PIWI-Interacting RNA: Its Biogenesis and Functions., Annu. Rev. Biochem (2015). doi: 10.1146/annurev-biochem-060614-034258. [DOI] [PubMed] [Google Scholar]
- [7].Jorjani H, Kehr S, Jedlinski DJ, Gumienny R, Hertel J, Stadler PF, Zavolan M, Gruber AR, An updated human snoRNAome, Nucleic Acids Res. (2016). doi: 10.1093/nar/gkw386. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [8].Rinn JL, Chang HY, Genome regulation by long noncoding RNAs., Annu. Rev. Biochem (2012). doi: 10.1146/annurev-biochem-051410-092902. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [9].Zhao Y, Li H, Fang S, Kang Y, Wu W, Hao Y, Li Z, Bu D, Sun N, Zhang MQ, Chen R, NONCODE 2016: An informative and valuable data source of long non-coding RNAs, Nucleic Acids Res. (2016). doi: 10.1093/nar/gkv1252. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [10].Jackson R, Kroehling L, Khitun A, Bailis W, Jarret A, York AG, Khan OM, Brewer JR, Skadow MH, Duizer C, Harman CCD, Chang L, Bielecki P, Solis AG, Steach HR, Slavoff S, Flavell RA, The translation of non-canonical open reading frames controls mucosal immunity, Nature. (2018). doi: 10.1038/s41586-018-0794-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [11].Matsumoto A, Pasut A, Matsumoto M, Yamashita R, Fung J, Monteleone E, Saghatelian A, Nakayama KI, Clohessy JG, Pandolfi PP, MTORC1 and muscle regeneration are regulated by the LINC00961-encoded SPAR polypeptide, Nature. (2017). doi: 10.1038/nature21034. [DOI] [PubMed] [Google Scholar]
- [12].Ji Z, Song R, Regev A, Struhl K, Many lncRNAs, 5′UTRs, and pseudogenes are translated and some are likely to express functional proteins, Elife. (2015). doi: 10.7554/eLife.08890. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [13].Candeias MM, Malbert-Colas L, Powell DJ, Daskalogianni C, Maslon MM, Naski N, Bourougaa K, Calvo F, Fåhraeus R, p53 mRNA controls p53 activity by managing Mdm2 functions, Nat. Cell Biol (2008). doi: 10.1038/ncb1770. [DOI] [PubMed] [Google Scholar]
- [14].Leygue E, Steroid receptor RNA activator (SRA1): unusual bifaceted gene products with suspected relevance to breast cancer., Nucl. Recept. Signal (2007). doi: 10.1621/nrs.05006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [15].Hubé F, Francastel C, Coding and non-coding RNAs, the frontier has never been so blurred, Front. Genet (2018). doi: 10.3389/fgene.2018.00140. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [16].Venkatraman A, He XC, Thorvaldsen JL, Sugimura R, Perry JM, Tao F, Zhao M, Christenson MK, Sanchez R, Yu JY, Peng L, Haug JS, Paulson A, Li H, Zhong XB, Clemens TL, Bartolomei MS, Li L, Maternal imprinting at the H19-Igf2 locus maintains adult haematopoietic stem cell quiescence, Nature. (2013). doi: 10.1038/nature12303. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [17].Imamura K, Imamachi N, Akizuki G, Kumakura M, Kawaguchi A, Nagata K, Kato A, Kawaguchi Y, Sato H, Yoneda M, Kai C, Yada T, Suzuki Y, Yamada T, Ozawa T, Kaneki K, Inoue T, Kobayashi M, Kodama T, Wada Y, Sekimizu K, Akimitsu N, Long Noncoding RNA NEAT1-Dependent SFPQ Relocation from Promoter Region to Paraspeckle Mediates IL8 Expression upon Immune Stimuli, Mol. Cell (2014). doi: 10.1016/j.molcel.2014.01.009. [DOI] [PubMed] [Google Scholar]
- [18].Collier SP, Henderson MA, Tossberg JT, Aune TM, Regulation of the Th1 Genomic Locus from Ifng through Tmevpg1 by T-bet, J. Immunol (2014). doi: 10.4049/jimmunol.1401099. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [19].Gomez JA, Wapinski OL, Yang YW, Bureau JF, Gopinath S, Monack DM, Chang HY, Brahic M, Kirkegaard K, The NeST long ncRNA controls microbial susceptibility and epigenetic activation of the interferon-γ locus, Cell. (2013). doi: 10.1016/j.cell.2013.01.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [20].Lewandowski JP, Lee JC, Hwang T, Sunwoo H, Goldstein JM, Groff AF, Chang N, Mallard W, Williams A, Henao-Meija J, Flavell RA, Lee JT, Gerhardinger C, Wagers AJ, Rinn JL, The Firre locus produces a trans-acting RNA molecule that functions in hematopoiesis, BioRxiv. (2019). doi: 10.1101/648279. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [21].Ouyang J, Zhu X, Chen Y, Wei H, Chen Q, Chi X, Qi B, Zhang L, Zhao Y, Gao GF, Wang G, Chen JL, NRAV, a long noncoding RNA, modulates antiviral responses through suppression of interferon-stimulated gene transcription, Cell Host Microbe. (2014). doi: 10.1016/j.chom.2014.10.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [22].Hu W, Yuan B, Flygare J, Lodish HF, Long noncoding RNA-mediated anti-apoptotic activity in murine erythroid terminal differentiation, Genes Dev. (2011). doi: 10.1101/gad.178780.111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [23].Zhang X, Lian Z, Padden C, Gerstein MB, Rozowsky J, Snyder M, Gingeras TR, Kapranov P, Weissman SM, Newburger PE, A myelopoiesis-associated regulatory intergenic noncoding RNA transcript within the human HOXA cluster, Blood. (2009). doi: 10.1182/blood-2008-06-162164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [24].Peng X, Gralinski L, Armour CD, Ferris MT, Thomas MJ, Proll S, Bradel-Tretheway BG, Korth MJ, Castle JC, Biery MC, Bouzek HK, Haynor DR, Frieman MB, Heise M, Raymond CK, Baric RS, Katze MG, Unique signatures Of long noncoding RNA expression in response to virus infection And altered innate immune signaling, MBio. (2010). doi: 10.1128/mBio.00206-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [25].Carpenter S, Aiello D, Atianand MK, Ricci EP, Gandhi P, Hall LL, Byron M, Monks B, Henry-Bezy M, Lawrence JB, O’Neill LAJ, Moore MJ, Caffrey DR, Fitzgerald KA, A long noncoding RNA mediates both activation and repression of immune response genes, Science (80-. ). (2013). doi: 10.1126/science.1240925. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [26].Li Z, Chao TC, Chang KY, Lin N, Patil VS, Shimizu C, Head SR, Burns JC, Rana TM, The long noncoding RNA THRIL regulates TNFα expression through its interaction with hnRNPL, Proc. Natl. Acad. Sci. U. S. A (2014). doi: 10.1073/pnas.1313768111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [27].Brazão TF, Johnson JS, Müller J, Heger A, Ponting CP, Tybulewicz VLJ, Long noncoding RNAs in B-cell development and activation, Blood. (2016). doi: 10.1182/blood-2015-11-680843. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [28].Vigneau S, Rohrlich P-S, Brahic M, Bureau J-F, Tmevpg1, a Candidate Gene for the Control of Theiler’s Virus Persistence, Could Be Implicated in the Regulation of Gamma Interferon, J. Virol (2003). doi: 10.1128/jvi.77.10.5632-5638.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [29].Atianand MK, Caffrey DR, Fitzgerald KA, Immunobiology of Long Noncoding RNAs, Annu. Rev. Immunol (2017). doi: 10.1146/annurev-immunol-041015-055459. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [30].Ranzani V, Rossetti G, Panzeri I, Arrigoni A, Bonnal RJP, Curti S, Gruarin P, Provasi E, Sugliano E, Marconi M, De Francesco R, Geginat J, Bodega B, Abrignani S, Pagani M, The long intergenic noncoding RNA landscape of human lymphocytes highlights the regulation of T cell differentiation by linc-MAF-4, Nat. Immunol (2015). doi: 10.1038/ni.3093. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [31].Mowel WK, Kotzin JJ, McCright SJ, Neal VD, Henao-Mejia J, Control of Immune Cell Homeostasis and Function by lncRNAs, Trends Immunol. (2018). doi: 10.1016/j.it.2017.08.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [32].Chen YG, Satpathy AT, Chang HY, Gene regulation in the immune system by long noncoding RNAs, Nat. Immunol (2017). doi: 10.1038/ni.3771. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [33].Derrien T, Johnson R, Bussotti G, Tanzer A, Djebali S, Tilgner H, Guernec G, Martin D, Merkel A, Knowles DG, Lagarde J, Veeravalli L, Ruan X, Ruan Y, Lassmann T, Carninci P, Brown JB, Lipovich L, Gonzalez JM, Thomas M, Davis CA, Shiekhattar R, Gingeras TR, Hubbard TJ, Notredame C, Harrow J, Guigó R, The GENCODE v7 catalog of human long noncoding RNAs: Analysis of their gene structure, evolution, and expression, Genome Res. (2012). doi: 10.1101/gr.132159.111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [34].Mattioli K, Volders P-J, Gerhardinger C, Lee JC, Maass PG, Melé M, Rinn JL, High-throughput functional analysis of lncRNA core promoters elucidates rules governing tissue specificity., Genome Res. (2019). doi: 10.1101/gr.242222.118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [35].Cabili M, Trapnell C, Goff L, Koziol M, Tazon-Vega B, Regev A, Rinn JL, Integrative annotation of human large intergenic noncoding RNAs reveals global properties and specific subclasses, Genes Dev. (2011). doi: 10.1101/gad.17446611. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [36].Marks LS, Bostwick DG, Prostate Cancer Specificity of PCA3 Gene Testing: Examples from Clinical Practice., Rev. Urol (2008). [PMC free article] [PubMed] [Google Scholar]
- [37].Hessels D, Schalken JA, The use of PCA3 in the diagnosis of prostate cancer, Nat. Rev. Urol (2009). doi: 10.1038/nrurol.2009.40. [DOI] [PubMed] [Google Scholar]
- [38].Meresse B, Malamut G, Cerf-Bensussan N, Celiac Disease: An Immunological Jigsaw, Immunity. (2012). doi: 10.1016/j.immuni.2012.06.006. [DOI] [PubMed] [Google Scholar]
- [39].Kagnoff MF, Celiac disease: Pathogenesis of a model immunogenetic disease, J. Clin. Invest (2007). doi: 10.1172/JCI30253. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [40].Dieli-Crimi R, Cénit MC, Núñez C, The genetics of celiac disease: A comprehensive review of clinical implications, J. Autoimmun (2015). doi: 10.1016/j.jaut.2015.07.003. [DOI] [PubMed] [Google Scholar]
- [41].Castellanos-Rubio A, Fernandez-Jimenez N, Kratchmarov R, Luo X, Bhagat G, Green PHR, Schneider R, Kiledjian M, Bilbao JR, Ghosh S, A long noncoding RNA associated with susceptibility to celiac disease, Science (80-.). (2016). doi: 10.1126/science.aad0467. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [42].Kotzin JJ, Spencer SP, McCright SJ, Kumar DBU, Collet MA, Mowel WK, Elliott EN, Uyar A, Makiya MA, Dunagin MC, Harman CCD, Virtue AT, Zhu S, Bailis W, Stein J, Hughes C, Raj A, Wherry EJ, Goff LA, Klion AD, Rinn JL, Williams A, Flavell RA, Henao-Mejia J, The long non-coding RNA Morrbid regulates Bim and short-lived myeloid cell lifespan, Nature. (2016). doi: 10.1038/nature19346. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [43].Corren J, Inhibition of interleukin-5 for the treatment of eosinophilic diseases., Discov. Med (2012). [PubMed] [Google Scholar]
- [44].Garrett JK, Jameson SC, Thomson B, Collins MH, Wagoner LE, Freese DK, Beck LA, Boyce JA, Filipovich AH, Villanueva JM, Sutton SA, Assa’ad AH, Rothenberg ME, Anti-interleukin-5 (mepolizumab) therapy for hypereosinophilic syndromes, J. Allergy Clin. Immunol (2004). doi: 10.1016/j.jaci.2003.10.049. [DOI] [PubMed] [Google Scholar]
- [45].Cai Z, Kotzin JJ, Ramdas B, Chen S, Nelanuthala S, Palam LR, Pandey R, Mali RS, Liu Y, Kelley MR, Sandusky G, Mohseni M, Williams A, Henao-Mejia J, Kapur R, Inhibition of Inflammatory Signaling in Tet2 Mutant Preleukemic Cells Mitigates Stress-Induced Abnormalities and Clonal Hematopoiesis, Cell Stem Cell. (2018). doi: 10.1016/j.stem.2018.10.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [46].Zhang F, Liu G, Wei C, Gao C, Hao J, Linc-MAF-4 regulates Th1/Th2 differentiation and is associated with the pathogenesis of multiple sclerosis by targeting MAF, FASEB J. (2017). doi: 10.1096/lj.201600838R. [DOI] [PubMed] [Google Scholar]
- [47].Lin PL, Flynn JAL, CD8 T cells and Mycobacterium tuberculosis infection, Semin. Immunopathol (2015). doi: 10.1007/s00281-015-0490-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [48].Agresta L, Hoebe KHN, Janssen EM, The emerging role of CD244 signaling in immune cells of the tumor microenvironment, Front. Immunol (2018). doi: 10.3389/fimmu.2018.02809. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [49].Wang Y, Zhong H, Xie X, Chen CY, Huang D, Shen L, Zhang H, Chen ZW, Zeng G, Long noncoding RNA derived from CD244 signaling epigenetically controls CD8+ T-cell immune responses in tuberculosis infection, Proc. Natl. Acad. Sci. U. S. A (2015). doi: 10.1073/pnas.1501662112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [50].Liu B, Sun L, Liu Q, Gong C, Yao Y, Lv X, Lin L, Yao H, Su F, Li D, Zeng M, Song E, A Cytoplasmic NF-κB Interacting Long Noncoding RNA Blocks IκB Phosphorylation and Suppresses Breast Cancer Metastasis, Cancer Cell. (2015). doi: 10.1016/j.ccell.2015.02.004. [DOI] [PubMed] [Google Scholar]
- [51].Huang D, Chen J, Yang L, Ouyang Q, Li J, Lao L, Zhao J, Liu J, Lu Y, Xing Y, Chen F, Su F, Yao H, Liu Q, Su S, Song E, NKILA lncRNA promotes tumor immune evasion by sensitizing T cells to activation-induced cell death, Nat. Immunol (2018). doi: 10.1038/s41590-018-0207-y. [DOI] [PubMed] [Google Scholar]
- [52].Lu Z, Li Y, Wang J, Che Y, Sun S, Huang J, Chen Z, He J, Long non-coding RNA NKILA inhibits migration and invasion of non-small cell lung cancer via NF-κB/Snail pathway, J. Exp. Clin. Cancer Res (2017). doi: 10.1186/s13046-017-0518-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [53].Yang T, Li S, Liu J, Yin D, Yang X, Tang Q, lncRNA-NKILA/NF-κB feedback loop modulates laryngeal cancer cell proliferation, invasion, and radioresistance, Cancer Med. (2018). doi: 10.1002/cam4.1405. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [54].Huang W, Cui X, Chen J, Feng Y, Song E, Li J, Liu Y, Long non-coding RNA NKILA inhibits migration and invasion of tongue squamous cell carcinoma cells via suppressing epithelialmesenchymal transition, Oncotarget. (2016). doi: 10.18632/oncotarget.11528. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [55].Zhang W, Guo Q, Liu G, Zheng F, Chen J, Huang D, Ding L, Yang X, Song E, Xiang Y, Yao H, NKILA represses nasopharyngeal carcinoma carcinogenesis and metastasis by NF-κB pathway inhibition, PLOS Genet. (2019). doi: 10.1371/journal.pgen.1008325. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [56].Lu B, Finn OJ, T-cell death and cancer immune tolerance, Cell Death Differ. (2008). doi: 10.1038/sj.cdd.4402274. [DOI] [PubMed] [Google Scholar]
- [57].Zhang DD, Wang WT, Xiong J, Xie XM, Cui SS, Zhao ZG, Li MJ, Zhang ZQ, Hao DL, Zhao X, Li YJ, Wang J, Chen HZ, Lv X, Liu DP, Long noncoding RNA LINC00305 promotes inflammation by activating the AHRR-NF-κ B pathway in human monocytes, Sci. Rep (2017). doi: 10.1038/srep46204. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [58].Lennox KA, Behlke MA, Cellular localization of long non-coding RNAs affects silencing by RNAi more than by antisense oligonucleotides, Nucleic Acids Res. (2016). doi: 10.1093/nar/gkv1206. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [59].Wittrup A, Lieberman J, Knocking down disease: A progress report on siRNA therapeutics, Nat. Rev. Genet (2015). doi: 10.1038/nrg3978. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [60].Hannon GJ, Rossi JJ, Unlocking the potential of the human genome with RNA interference, Nature. (2004). doi: 10.1038/nature02870. [DOI] [PubMed] [Google Scholar]
- [61].Vickers TA, Koo S, Bennett CF, Crooke ST, Dean NM, Baker BF, Efficient reduction of target RNAs by small interfering RNA and RNase H-dependent antisense agents. A comparative analysis, J. Biol. Chem (2003). doi: 10.1074/jbc.M210326200. [DOI] [PubMed] [Google Scholar]
- [62].Akinc A, Querbes W, De S, Qin J, Frank-Kamenetsky M, Jayaprakash KN, Jayaraman M, Rajeev KG, Cantley WL, Dorkin JR, Butler JS, Qin L, Racie T, Sprague A, Fava E, Zeigerer A, Hope MJ, Zerial M, Sah DW, Fitzgerald K, Tracy MA, Manoharan M, Koteliansky V, De Fougerolles A, Maier MA, Targeted delivery of RNAi therapeutics with endogenous and exogenous ligand-based mechanisms, Mol. Ther (2010). doi: 10.1038/mt.2010.85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [63].Springer AD, Dowdy SF, GalNAc-siRNA Conjugates: Leading the Way for Delivery of RNAi Therapeutics, Nucleic Acid Ther. (2018). doi: 10.1089/nat.2018.0736. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [64].Rozema DB, Lewis DL, Wakefield DH, Wong SC, Klein JJ, Roesch PL, Bertin SL, Reppen TW, Chu Q, Blokhin AV, Hagstrom JE, Wolff JA, Dynamic PolyConjugates for targeted in vivo delivery of siRNA to hepatocytes, Proc. Natl. Acad. Sci. U. S. A (2007). doi: 10.1073/pnas.0703778104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [65].Adams D, Gonzalez-Duarte A, O’Riordan WD, Yang CC, Ueda M, Kristen AV, Tournev I, Schmidt HH, Coelho T, Berk JL, Lin KP, Vita G, Attarian S, Planté-Bordeneuve V, Mezei MM, Campistol JM, Buades J, Brannagan TH, Kim BJ, Oh J, Parman Y, Sekijima Y, Hawkins PN, Solomon SD, Polydefkis M, Dyck PJ, Gandhi PJ, Goyal S, Chen J, Strahs AL, Nochur SV, Sweetser MT, Garg PP, Vaishnaw AK, Gollob JA, Suhr OB, Patisiran, an RNAi therapeutic, for hereditary transthyretin amyloidosis, N. Engl. J. Med (2018). doi: 10.1056/NEJMoa1716153. [DOI] [PubMed] [Google Scholar]
- [66].Gupta RA, Shah N, Wang KC, Kim J, Horlings HM, Wong DJ, Tsai MC, Hung T, Argani P, Rinn JL, Wang Y, Brzoska P, Kong B, Li R, West RB, Van De Vijver MJ, Sukumar S, Chang HY, Long non-coding RNA HOTAIR reprograms chromatin state to promote cancer metastasis, Nature. (2010). doi: 10.1038/nature08975. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [67].Ren S, Liu Y, Xu W, Sun Y, Lu J, Wang F, Wei M, Shen J, Hou J, Gao X, Xu C, Huang J, Zhao Y, Sun Y, Long noncoding RNA MALAT-1 is a new potential therapeutic target for castration resistant prostate cancer, J. Urol (2013). doi: 10.1016/j.juro.2013.07.001. [DOI] [PubMed] [Google Scholar]
- [68].Kurreck J, Design of antisense oligonucleotides stabilized by locked nucleic acids, Nucleic Acids Res. (2002). doi: 10.1093/nar/30.9.1911. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [69].Veedu RN, Wengel J, Locked nucleic acids: Promising nucleic acid analogs for therapeutic applications, Chem. Biodivers (2010). doi: 10.1002/cbdv.200900343. [DOI] [PubMed] [Google Scholar]
- [70].Castanotto D, Lin M, Kowolik C, Wang LA, Ren XQ, Soifer HS, Koch T, Hansen BR, Oerum H, Armstrong B, Wang Z, Bauer P, Rossi J, Stein CA, A cytoplasmic pathway for gapmer antisense oligonucleotide-mediated gene silencing in mammalian cells, Nucleic Acids Res. (2015). doi: 10.1093/nar/gkv964. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [71].Vickers TA, Crooke ST, The rates of the major steps in the molecular mechanism of RNase H1-dependent antisense oligonucleotide induced degradation of RNA, Nucleic Acids Res. (2015). doi: 10.1093/nar/gkv920. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [72].Shen X, Corey DR, Chemistry, mechanism and clinical status of antisense oligonucleotides and duplex RNAs, Nucleic Acids Res. (2018). doi: 10.1093/nar/gkx1239. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [73].Paton DM, Nusinersen: Antisense oligonucleotide to increase SMN protein production in spinal muscular atrophy, Drugs of Today. (2017). doi: 10.1358/dot.2017.53.6.2652413. [DOI] [PubMed] [Google Scholar]
- [74].Finkel RS, Chiriboga CA, Vajsar J, Day JW, Montes J, De Vivo DC, Yamashita M, Rigo F, Hung G, Schneider E, Norris DA, Xia S, Bennett CF, Bishop KM, Treatment of infantile-onset spinal muscular atrophy with nusinersen: a phase 2, open-label, dose-escalation study, Lancet. (2016). doi: 10.1016/S0140-6736(16)31408-8. [DOI] [PubMed] [Google Scholar]
- [75].Hung G, Xiao X, Peralta R, Bhattacharjee G, Murray S, Norris D, Guo S, Monia BP, Characterization of target mRNA reduction through in situ RNA hybridization in multiple organ systems following systemic antisense treatment in animals, Nucleic Acid Ther. (2013). doi: 10.1089/nat.2013.0443. [DOI] [PubMed] [Google Scholar]
- [76].Crooke ST, Baker BF, Pham NC, Hughes SG, Kwoh TJ, Cai D, Tsimikas S, Geary RS, Bhanot S, The effects of 2’-o-methoxyethyl oligonucleotides on renal function in humans, Nucleic Acid Ther. (2018). doi: 10.1089/nat.2017.0693. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [77].Henry SP, Kim TW, Kramer-Stickland K, Zanardi TA, Fey RA, Levin AA, Toxicologic properties of 2′-o-methoxyethyl chimeric antisense inhibitors in animals and man, in: Antisense Drug Technol. Princ. Strateg. Appl. Second Ed, 2007. doi: 10.1201/9780849387951.ch12. [DOI] [Google Scholar]
- [78].Geary RS, Norris D, Yu R, Bennett CF, Pharmacokinetics, biodistribution and cell uptake of antisense oligonucleotides, Adv. Drug Deliv. Rev (2015). doi: 10.1016/j.addr.2015.01.008. [DOI] [PubMed] [Google Scholar]
- [79].Ämmälä C, Drury WJ, Knerr L, Ahlstedt L, Stillemark-Billton P, Wennberg-Huldt C, Andersson EM, Valeur E, Jansson-Löfmark R, Janzén D, Sundström L, Meuller J, Claesson J, Andersson P, Johansson C, Lee RG, Prakash TP, Seth PP, Monia BP, Andersson S, Targeted delivery of antisense oligonucleotides to pancreatic β-cells, Sci. Adv (2018). doi: 10.1126/sciadv.aat3386. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [80].Scharner J, Qi S, Rigo F, Bennett CF, Krainer AR, Delivery of GalNAc-Conjugated Splice-Switching ASOs to Non-hepatic Cells through Ectopic Expression of Asialoglycoprotein Receptor, Mol. Ther. - Nucleic Acids (2019). doi: 10.1016/j.omtn.2019.02.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [81].Alarcón-Arís D, Recasens A, Galofré M, Carballo-Carbajal E, Zacchi N, Ruiz-Bronchal E, Pavia-Collado R, Chica R, Ferres-Coy A, Santos M, Revilla R, Montefeltro A, Fariñas L, Artigas F, Vila M, Bortolozzi A, Selective α-Synuclein Knockdown in Monoamine Neurons by Intranasal Oligonucleotide Delivery: Potential Therapy for Parkinson’s Disease, Mol. Ther (2018). doi: 10.1016/j.ymthe.2017.11.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [82].Prakash TP, Graham MJ, Yu J, Carty R, Low A, Chappell A, Schmidt K, Zhao C, Aghajan M, Murray HF, Riney S, Booten SL, Murray SF, Gaus H, Crosby J, Lima WF, Guo S, Monia BP, Swayze EE, Seth PP, Targeted delivery of antisense oligonucleotides to hepatocytes using triantennary N-acetyl galactosamine improves potency 10-fold in mice, Nucleic Acids Res. (2014). doi: 10.1093/nar/gku531. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [83].Arun G, Diermeier S, Akerman M, Chang KC, Wilkinson JE, Heam S, Kim Y, MacLeod AR, Krainer AR, Norton L, Brogi E, Egeblad M, Spector DL, Differentiation of mammary tumors and reduction in metastasis upon Malat1 lncRNA loss, Genes Dev. (2016). doi: 10.1101/gad.270959.115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [84].Hu Q, Ye Y, Chan LC, Li Y, Liang K, Lin A, Egranov SD, Zhang Y, Xia W, Gong J, Pan Y, Chatterjee SS, Yao J, Evans KW, Nguyen TK, Park PK, Liu J, Coarfa C, Donepudi SR, Putluri V, Putluri N, Sreekumar A, Ambati CR, Hawke DH, Marks JR, Gunaratne PH, Caudle AS, Sahin AA, Hortobagyi GN, Meric-Bernstam F, Chen L, Yu D, Hung MC, Curran MA, Han L, Lin C, Yang L, Oncogenic lncRNA downregulates cancer cell antigen presentation and intrinsic tumor suppression, Nat. Immunol (2019). doi: 10.1038/s41590-019-0400-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [85].Garchow BG, Encinas OB, Leung YT, Tsao PY, Eisenberg RA, Caricchio R, Obad S, Petri A, Kauppinen S, Kiriakidou M, Silencing of microR6-21 in vivo ameliorates autoimmune splenomegaly in lupus mice, EMBO Mol. Med (2011). doi: 10.1002/emmm.201100171. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [86].Uemura Y, Hagiwara K, Kobayashi K, The intratracheal administration of locked nucleic acid containing antisense oligonucleotides induced gene silencing and an immune-stimulatory effect in the murine lung, PLoS One. (2017). doi: 10.1371/journal.pone.0187286. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [87].Uemura Y, Kobayashi K, Targeting murine alveolar macrophages by the intratracheal administration of locked nucleic acid containing antisense oligonucleotides, Drug Deliv. (2019). doi: 10.1080/10717544.2019.1648589. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [88].Murugaiyan G, Beynon V, Mittal A, Joller N, Weiner HL, Silencing MicroRNA-155 Ameliorates Experimental Autoimmune Encephalomyelitis, J. Immunol (2011). doi: 10.4049/jimmunol.1003952. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [89].Nakamura H, Oda Y, Iwai S, Inoue H, Ohtsuka E, Kanaya S, Kimura S, Katsuda C, Katayanagi K, Morikawa K, Miyashiro H, Ikehara M, How does RNase H recognize a DNA·RNA hybrid?, Proc. Natl. Acad. Sci. U. S. A (1991). doi: 10.1073/pnas.88.24.11535. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [90].Shimojo M, Kasahara Y, Inoue M, ichi Tsunoda S, Shudo Y, Kurata T, Obika S, A gapmer antisense oligonucleotide targeting SRRM4 is a novel therapeutic medicine for lung cancer, Sci. Rep (2019). doi: 10.1038/s41598-019-43100-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [91].Benson MD, Pandey S, Witchell D, Jazayeri A, Siwkowski A, Monia B, Kluve-Beckerman B, Antisense oligonucleotide therapy for TTR amyloidosis, Amyloid. (2011). doi: 10.3109/13506129.2011.574354. [DOI] [PubMed] [Google Scholar]
- [92].Benson MD, Waddington-Cruz M, Berk JL, Polydefkis M, Dyck PJ, Wang AK, Planté-Bordeneuve V, Barroso FA, Merlini G, Obici L, Scheinberg M, Brannagan TH, Litchy WJ, Whelan C, Drachman BM, Adams D, Heitner SB, Conceição I, Schmidt HH, Vita G, Campistol JM, Gamez J, Gorevic PD, Gane E, Shah AM, Solomon SD, Monia BP, Hughes SG, Jesse Kwoh T, McEvoy BW, Jung SW, Baker BF, Ackermann EJ, Gertz MA, Coelho T, Inotersen treatment for patients with Hereditary transthyretin amyloidosis, N. Engl. J. Med (2018). doi: 10.1056/NEJMoa1716793. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [93].Gertz MA, Scheinberg M, Waddington-Cruz M, Heitner SB, Karam C, Drachman B, Khella S, Whelan C, Obici L, Inotersen for the treatment of adults with polyneuropathy caused by hereditary transthyretin-mediated amyloidosis, Expert Rev. Clin. Pharmacol (2019). doi: 10.1080/17512433.2019.1635008. [DOI] [PubMed] [Google Scholar]
- [94].Keam SJ, Inotersen: First Global Approval, Drugs. (2018). doi: 10.1007/s40265-018-0968-5. [DOI] [PubMed] [Google Scholar]
- [95].Blum M, De Robertis EM, Wallingford JB, Niehrs C, Morpholinos: Antisense and Sensibility, Dev. Cell (2015). doi: 10.1016/j.devcel.2015.09.017. [DOI] [PubMed] [Google Scholar]
- [96].Summerton J, Morpholino antisense oligomers: The case for an RNase H-independent structural type, Biochim. Biophys. Acta - Gene Struct. Expr (1999). doi: 10.1016/S0167-4781(99)00150-5. [DOI] [PubMed] [Google Scholar]
- [97].Alter J, Lou F, Rabinowitz A, Yin HF, Rosenfeld J, Wilton SD, Partridge TA, Qi LL, Systemic delivery of morpholino oligonucleotide restores dystrophin expression bodywide and improves dystrophic pathology, Nat. Med (2006). doi: 10.1038/nm1345. [DOI] [PubMed] [Google Scholar]
- [98].Wu JC, Meng QC, Ren HM, Wang HT, Wu J, Wang Q, Recent advances in peptide nucleic acid for cancer bionanotechnology, Acta Pharmacol. Sin (2017). doi: 10.1038/aps.2017.33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [99].Özeş AR, Wang Y, Zong X, Fang F, Pilrose J, Nephew KP, Therapeutic targeting using tumor specific peptides inhibits long non-coding RNA HOTAIR activity in ovarian and breast cancer, Sci. Rep (2017). doi: 10.1038/s41598-017-00966-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [100].Pedram Fatemi R, Salah-Uddin S, Modarresi F, Khoury N, Wahlestedt C, Faghihi MA, Screening for small-molecule modulators of long noncoding RNA-protein interactions using alphascreen, J. Biomol. Screen (2015). doi: 10.1177/1087057115594187. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [101].Shi Y, Parag S, Patel R, Lui A, Murr M, Cai J, Patel NA, Stabilization of lncRNA GAS5 by a Small Molecule and Its Implications in Diabetic Adipocytes, Cell Chem. Biol (2019). doi: 10.1016/j.chembiol.2018.11.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [102].Abulwerdi FA, Xu W, Ageeli AA, Yonkunas MJ, Arun G, Nam H, Schneekloth JS, Dayie TK, Spector D, Baird N, Le Grice SFJ, Selective Small-Molecule Targeting of a Triple Helix Encoded by the Long Noncoding RNA, MALAT1, ACS Chem. Biol (2019). doi: 10.1021/acschembio.8b00807. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [103].Doudna JA, Charpentier E, The new frontier of genome engineering with CRISPR-Cas9, Science (80-. ). (2014). doi: 10.1126/science.1258096. [DOI] [PubMed] [Google Scholar]
- [104].Wang H, La Russa M, Qi LS, CRISPR/Cas9 in Genome Editing and Beyond, Annu. Rev. Biochem (2016). doi: 10.1146/annurev-biochem-060815-014607. [DOI] [PubMed] [Google Scholar]
- [105].Kotzin JJ, Iseka F, Wright J, Basavappa MG, Clark ML, Ali MA, Abdel-Hakeem MS, Robertson TF, Mowel WK, Joannas L, Neal VD, Spencer SP, Syrett CM, Anguera MC, Williams A, Wherry EJ, Henao-Mejia J, The long noncoding RNA Morrbid regulates CD8 T cells in response to viral infection, Proc. Natl. Acad. Sci. U. S. A (2019). doi: 10.1073/pnas.1819457116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [106].Mowel WK, McCright SJ, Kotzin JJ, Collet MA, Uyar A, Chen X, DeLaney A, Spencer SP, Virtue AT, Yang EJ, Villarino A, Kurachi M, Dunagin MC, Pritchard GH, Stein J, Hughes C, Fonseca-Pereira D, Veiga-Fernandes H, Raj A, Kambayashi T, Brodsky IE, O’Shea JJ, Wherry EJ, Goff LA, Rinn JL, Williams A, Flavell RA, Henao-Mejia J, Group 1 Innate Lymphoid Cell Lineage Identity Is Determined by a cis-Regulatory Element Marked by a Long Non-coding RNA, Immunity. (2017). doi: 10.1016/j.immuni.2017.08.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [107].Li G, Ruan X, Auerbach RK, Sandhu KS, Zheng M, Wang P, Poh HM, Goh Y, Lim J, Zhang J, Sim HS, Peh SQ, Mulawadi FH, Ong CT, Orlov YL, Hong S, Zhang Z, Landt S, Raha D, Euskirchen G, Wei CL, Ge W, Wang H, Davis C, Fisher-Aylor KI, Mortazavi A, Gerstein M, Gingeras T, Wold B, Sun Y, Fullwood MJ, Cheung E, Liu E, Sung WK, Snyder M, Ruan Y, Extensive promoter-centered chromatin interactions provide a topological basis for transcription regulation, Cell. (2012). doi: 10.1016/j.cell.2011.12.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [108].Mifsud B, Tavares-Cadete F, Young AN, Sugar R, Schoenfelder S, Ferreira L, Wingett SW, Andrews S, Grey W, Ewels PA, Herman B, Happe S, Higgs A, Leproust E, Follows GA, Fraser P, Luscombe NM, Osborne CS, Mapping long-range promoter contacts in human cells with high-resolution capture Hi-C, Nat. Genet (2015). doi: 10.1038/ng.3286. [DOI] [PubMed] [Google Scholar]
- [109].Sahlén P, Abdullayev I, Ramsköld D, Matskova L, Rilakovic N, Lötstedt B, Albert TJ, Lundeberg J, Sandberg R, Genome-wide mapping of promoter-anchored interactions with close to single-enhancer resolution, Genome Biol. (2015). doi: 10.1186/s13059-015-0727-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [110].Liu SJ, Horlbeck MA, Cho SW, Birk HS, Malatesta M, He D, Attenello FJ, Villalta JE, Cho MY, Chen Y, Mandegar MA, Olvera MP, Gilbert LA, Conklin BR, Chang HY, Weissman JS, Lim DA, CRISPRi-based genome-scale identification of functional long noncoding RNA loci in human cells, Science (80-. ). (2017). doi: 10.1126/science.aah7111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [111].Konermann S, Lotfy P, Brideau NJ, Oki J, Shokhirev MN, Hsu PD, Transcriptome Engineering with RNA-Targeting Type VI-D CRISPR Effectors, Cell. (2018). doi: 10.1016/j.cell.2018.02.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [112].Austin PJ, Tsitsiou E, Boardman C, Jones SW, Lindsay MA, Adcock IM, Chung KF, Perry MM, Transcriptional profiling identifies the long noncoding RNA plasmacytoma variant translocation (PVT1) as a novel regulator of the asthmatic phenotype in human airway smooth muscle, J. Allergy Clin. Immunol (2017). doi: 10.1016/j.jaci.2016.06.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [113].Zhu Y, Mao D, Gao W, Han G, Hu H, Analysis of lncRNA expression in patients with eosinophilic and neutrophilic asthma focusing on LNC-000127, Front. Genet (2019). doi: 10.3389/fgene.2019.00141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [114].Sherrill JD, Kiran KC, Blanchard C, Stucke EM, Kemme KA, Collins MH, Abonia JP, Putnam PE, Mukkada VA, Kaul A, Kocoshis SA, Kushner JP, Plassard AJ, Karns RA, Dexheimer PJ, Aronow BJ, Rothenberg ME, Analysis and expansion of the eosinophilic esophagitis transcriptome by RNA sequencing, Genes Immun. (2014). doi: 10.1038/gene.2014.27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [115].Mayama T, Marr AK, Kino T, Differential Expression of Glucocorticoid Receptor Noncoding RNA Repressor Gas5 in Autoimmune and Inflammatory Diseases, Horm. Metab. Res (2016). doi: 10.1055/s-0042-106898. [DOI] [PubMed] [Google Scholar]
- [116].Lucafò M, Di Silvestre A, Romano M, Avian A, Antonelli R, Martelossi S, Naviglio S, Tommasini A, Stocco G, Ventura A, Decorti G, De Iudicibus S, Role of the Long Non-Coding RNA Growth Arrest-Specific 5 in Glucocorticoid Response in Children with Inflammatory Bowel Disease, Basic Clin. Pharmacol. Toxicol (2018). doi: 10.1111/bcpt.12851. [DOI] [PubMed] [Google Scholar]
- [117].Song J, Kim D, Han J, Kim Y, Lee M, Jin EJ, PBMC and exosome-derived Hotair is a critical regulator and potent marker for rheumatoid arthritis, Clin. Exp. Med (2014). doi: 10.1007/s10238-013-0271-4. [DOI] [PubMed] [Google Scholar]
- [118].Stuhlmüller B, Kunisch E, Franz J, Martinez-Gamboa L, Hernandez MM, Pruss A, Ulbrich N, Erdmann VA, Burmester GR, Kinnet RW, Detection of oncofetal H19 RNA in rheumatoid arthritis synovial tissue, Am. J. Pathol (2003). doi: 10.1016/S0002-9440(10)63450-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [119].Chen SW, Wang PY, Liu YC, Sun L, Zhu J, Zuo S, Ma J, Li TY, Zhang JL, Chen GW, Wang X, Zhu QR, Zheng YW, Chen ZY, Yao ZH, Pan YS, Effect of Long Noncoding RNA H19 Overexpression on Intestinal Barrier Function and Its Potential Role in the Pathogenesis of Ulcerative Colitis, Inflamm. Bowel Dis (2016). doi: 10.1097/MIB.0000000000000932. [DOI] [PubMed] [Google Scholar]
- [120].Zhang F, Wu L, Qian J, Qu B, Xia S, La T, Wu Y, Ma J, Zeng J, Guo Q, Cui Y, Yang W, Huang J, Zhu W, Yao Y, Shen N, Tang Y, Identification of the long noncoding RNA NEAT1 as a novel inflammatory regulator acting through MAPK pathway in human lupus, J. Autoimmun (2016). doi: 10.1016/j.jaut.2016.07.012. [DOI] [PubMed] [Google Scholar]
- [121].Wu Y, Zhang F, Ma J, Zhang X, Wu L, Qu B, Xia S, Chen S, Tang Y, Shen N, Association of large intergenic noncoding RNA expression with disease activity and organ damage in systemic lupus erythematosus, Arthritis Res. Ther (2015). doi: 10.1186/s13075-015-0632-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [122].Sonkoly E, Bata-Csorgo Z, Pivarcsi A, Polyanka H, Kenderessy-Szabo A, Molnar G, Szentpali K, Bari L, Megyeri K, Mandi Y, Dobozy A, Kemeny L, Szell M, Identification and characterization of a novel, psoriasis susceptibility-related noncoding RNA gene, PRINS, J. Biol. Chem (2005). doi: 10.1074/jbc.M501704200. [DOI] [PubMed] [Google Scholar]
- [123].Szegedi K, Sonkoly E, Nagy N, Németh IB, Bata-Csörgo Z, Kemény L, Dobozy A, Széll M, The anti-apoptotic protein G1P3 is overexpressed in psoriasis and regulated by the non-coding RNA, PRINS, Exp. Dermatol (2010). doi: 10.1111/j.1600-0625.2010.01066.x. [DOI] [PubMed] [Google Scholar]
- [124].Szegedi K, Göblös A, Bacsa S, Antal M, Németh IB, Bata-Csörgo Z, Kemény L, Dobozy A, Széll M, Expression and functional studies on the noncoding RNA, PRINS, Int. J. Mol. Sci (2013). doi: 10.3390/ijms14010205. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [125].Holm SJ, Sánchez F, Carlén LM, Mallbris L, Ståhle M, O’Brien KP, HLA-Cw*0602 associates more strongly to psoriasis in the Swedish population than variants of the novel 6p21.3 gene PSORS1C3, Acta Derm. Venereol (2005). doi: 10.1080/00015550410023527. [DOI] [PubMed] [Google Scholar]
- [126].Padua D, Mahurkar-Joshi S, Law IKM, Polytarchou C, Vu JP, Pisegna JR, Shih D, Iliopoulos D, Pothoulakis C, A long noncoding RNA signature for ulcerative colitis identifies IFNG-AS1 as an enhancer of inflammation, Am. J. Physiol. - Gastrointest. Liver Physiol (2016). doi: 10.1152/ajpgi.00212.2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [127].Rankin CR, Lokhandwala ZA, Huang R, Pekow J, Pothoulakis C, Padua D, Linear and circular CDKN2B-AS1 expression is associated with Inflammatory Bowel Disease and participates in intestinal barrier formation, Life Sci. (2019). doi: 10.1016/j.lfs.2019.116571. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [128].Collier SP, Collins PL, Williams CL, Boothby MR, Aune TM, Cutting Edge: Influence of Tmevpg1, a Long Intergenic Noncoding RNA, on the Expression of Ifng by Th1 Cells, J. Immunol (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [129].Peng H, Liu Y, Tian J, Ma J, Tang X, Rui K, Tian X, Mao C, Lu L, Xu H, Jiang P, Wang S, The Long Noncoding RNA IFNG-AS1 Promotes T Helper Type 1 Cells Response in Patients with Hashimoto’s Thyroiditis, Sci. Rep (2015). doi: 10.1038/srep17702. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [130].Zemmour D, Pratama A, Loughhead SM, Mathis Di., Benoist C, Flicr, a long noncoding RNA, modulates Foxp3 expression and autoimmunity, Proc. Natl. Acad. Sci. U. S. A (2017). doi: 10.1073/pnas.1700946114. [DOI] [PMC free article] [PubMed] [Google Scholar]

