REVIEW ARTICLE |
https://doi.org/10.5005/jp-journals-11002-0012 |
Non-coding RNAs in Neonatal Necrotizing Enterocolitis
1,2Department of Pediatrics, University of South Florida Health Morsani College of Medicine, Tampa, Florida, United States of America
3Global Newborn Society, Clarksville, Maryland, United States of America
Corresponding Author: Akhil Maheshwari, Global Newborn Society, Clarksville, Maryland, United States of America, Phone: +1 708 910 8729, e-mail: akhil1uic@gmail.com
How to cite this article: Donda K, Torres BA, Maheshwari A. Non-coding RNAs in Neonatal Necrotizing Enterocolitis. Newborn 2022;1(1):120–130.
Source of support: NIH awards HL133022 and HL124078 (to AM)
Conflict of interest: None
ABSTRACT
The incomplete understanding of the etiopathogenesis of necrotizing enterocolitis (NEC) contributes to the lack of timely diagnosis and limited therapeutic options. Non-coding RNAs (ncRNAs) have emerged as key regulators of gene expression in various pathways that can modulate various physiological and pathological processes. Despite several studies revealing the role of ncRNAs in intestinal inflammatory diseases in adults, these remain largely unexplored in NEC. In this article, we review the information on ncRNAs that have been specifically identified in NEC or have been noted in other inflammatory bowel disorders that share some of the histopathological abnormalities seen frequently in NEC. We have assimilated the most current research findings on ncRNAs in intestinal diseases. This is an attempt to explore a novel field that has immense potential for future translational and clinical research in preventing, detecting, and treating NEC.
Keywords: Genetic predisposition, Intestinal inflammation, Necrotizing enterocolitis, Neonates, Non-coding RNA, Spontaneous intestinal perforation.
IMPACT
Current information categorizes NEC as a multifactorial, inflammatory bowel necrosis of newborn infants.
Non-coding RNAs (ncRNAs) may influence the risk of occurrence of NEC.
ncRNAs may modulate the severity of intestinal injury and consequently the clinical outcome of NEC.
ncRNAs have been linked with inflammatory intestinal diseases of adults that share histopathological findings with neonatal NEC and, hence, need to be explored.
INTRODUCTION
Necrotizing enterocolitis (NEC), an inflammatory necrosis that may involve parts of the small and the large intestine, is one of the most common and serious diseases in premature infants causing significant morbidity and mortality. The etiopathogenesis of NEC in neonates is multifactorial. Prematurity is the prime risk factor for NEC development. In addition, various prenatal and postnatal factors contribute to the disease development and progression such as antenatal steroids, type of feeding, gut dysbiosis, hypoxic-ischemic injury, severe anemia requiring packed red cell transfusion. Clinically, the presenting features include abdominal distension, hematochezia, emesis, and feeding intolerance, which can be associated with subtle changes in vital signs including temperature instability, tachycardia, and lethargy. Abdominal radiography remains the diagnostic tool of choice with pathognomonic sign of pneumatosis intestinalis. The disease usually involves ileocolic region and colon; histopathologically, NEC is characterized by exaggerated inflammation, coagulative necrosis, pneumatosis intestinalis, intestinal hemorrhage, and reparative changes.1 The treatment is currently limited to supportive care in an attempt to prevent further injury to the intestine. Despite major advances in neonatology, the options to diagnose and treat NEC are few, and the available strategies have not made a significant impact bringing down the prevalence and improving outcomes. There is a need for more research to explore novel biomarkers and potential therapeutic targets.
Recently, rapidly growing interest in genetic research along with the availability of in-depth transcriptome sequencing techniques has exponentially expanded our understanding of gene expression and its regulation. This added knowledge has introduced the possibility of a complex interaction between clinical risk factors and genetic susceptibility explaining inter-individual variability of NEC susceptibility, progression, and prognosis. Non-coding RNAs (ncRNAs) have been unraveled recently as one of the key regulators of gene expression. In this review, we aim to provide currently available evidence from human and animal studies on role of ncRNAs in the pathogenesis of NEC. We also present the evidence for ncRNAs in other intestinal diseases that share similar histopathological characteristics with NEC for future direction. We have extensively searched in the databases PubMed, EMBASE, and Scopus after short-listing the keywords to describe the histopathological and clinical features of NEC.
NON-CODING RNAS
The ncRNAs, as the name suggests, are RNA molecules that are not translated into proteins. Since first discovered in eukaryotic cells in 1989, ncRNAs have gained tremendous visibility. About 80–90% of living cell genome is transcribed, however, only less than 2% of that transcribed RNA encodes for protein. Thus, RNAs can be categorized into coding and ncRNAs. ncRNA molecules are further categorized based on function into (1) housekeeping ncRNAs, such as transfer RNAs (tRNAs), ribosomal RNAs (rRNAs), small nuclear RNAs (snRNAs), and small nucleolar RNAs (snoRNAs), and (2) regulatory ncRNAs. The ncRNAs can also be categorized into two groups based on their nucleotide size, (1) small ncRNAs (<200 nucleotides), and (2) long ncRNAs (>200 nucleotides). The most studied small ncRNAs are <50 nucleotides long and therefore, to better categorize 50–200 nucleotide-long ncRNAS, a term “mid-size” ncRNAs have been proposed which includes snoRNAs, promoter-associated small RNAs (PASRs), transcription start site-associated RNAs (TSSa-RNAs), and promoter upstream transcripts (PROMPTs).2,3 Circular RNAs (circ-RNAs) are another variant, which are comprised of a covalently closed continuous loop that lacks the 5' cap and the 3' tail.4 Similarly, pyknons are recognizable non-random sequences that may be repeated mainly in the non-coding genomic DNA.5 Different types of ncRNAs are depicted in Figure 1. So far, microRNAs (miRNAs), piwi-interacting RNAs (piRNAs), small interfering RNAs (siRNAs), lncRNAs, and circular RNAs (circRNAs) have been studied.
Fig. 1: Classification of non-coding RNAs. ceRNA, competing endogenous RNA; cisRNA, cis-acting RNA; crasiRNA, centromere repeat-associated small interacting RNA; miRNA, microRNA; ncRNA, non-coding RNA; PASR, promoter-associated small RNA; piRNA, piwi-interacting RNA; PROMPT, promoter upstream transcripts; rRNA, ribosomal RNA; siRNA, small interfering RNA; snRNA, small nuclear RNA; snoRNA, small nucleolar RNA; tRNA, transfer RNA; telsRNA, telomere-specific small RNA; transRNA, trans-acting RNA; TSSa-RNA, transcription start site-associated RNAs
MicroRNAs
MicroRNAs (miRNAs) are endogenous, conserved, 21–23 nucleotide-long ncRNAs involved in posttranscriptional silencing of gene expression.6 Approximately 1–3% of the mammalian genome is now known to code for miRNAs. MiRNA genes are distributed throughout the genome and can be seen in intronic sequences of protein-coding genes, within intronic or exonic regions of ncRNAs, and even between independent transcription units (intergenic). MiRNAs may carry specific promoters for independent transcription, share promoters with host genes, or could be cotranscribed as a single primary miRNA transcript.7
The DNA sequences encoding for mRNAs are first transcribed in the nucleus by the RNA polymerase II-producing primary RNAs (pri-miRNAs). The pri-miRNA is then processed in a stepwise manner by nuclear as well as cytoplasmic endoribonucleases forming mature miRNA. Drosha, a type III ribonuclease located in the nucleus, processes pri-miRNA into ~70 nucleotide-containing precursor (pre-) miRNAs. These oligonucleotides are then translocated into the cytoplasm by the exportin-5 shuttle.8 In the cytoplasm, this pre-miRNA is further processed by another type III ribonuclease, dicer, into a mature miRNA. The 3'-end of the miRNA binds the Argonaute protein in a specialized oligonucleotide/oligosaccharide-binding fold to form an RNA-induced silencing complex (RISC).9 RISCs bind approximately complementary sequences in the 3'-untranslated region (UTR)s of target mRNAs and regulate protein output by either promoting mRNA degradation and/or inhibiting translation.10 Genomic analyses of miRNA-target interactions show conserved complementarity for approximately 6–8 base pairs from position II of the miRNA. This region (nucleotides 2–7 at the 5' end of the miRNA) is often termed the “seed” sequence for computational miRNA target prediction.11
The function of most miRNAs is still unclear. A single miRNA can regulate hundreds of genes, because only a few RNA nucleotides (2 through 7 or 8) are needed to recruit RISC and bind the seed sequence of a target mRNA for repression.12,13 Many miRNAs are now believed to modulate cellular differentiation, proliferation, apoptosis, inflammation, and stem cell maintenance and may also indicate the timing of various events during development.6 These features, together with the observation that miRNAs can be secreted and stay stable in plasma, make them prominent, accessible biomarkers as well as therapeutic targets.14
Piwi-interacting RNAs
Piwi-interacting RNAs (piRNAs) are 26–31 nucleotide-long ncRNAs that interact with the piwi family of proteins. The transcription process of piRNA is dicer-independent and is activated in the piRNA gene clusters on heterochromatin. Pre-initiation complex (PIC) is formed after recruitment of RNA polymerase II, and other transcription factors that in turn initiate piRNA transcription and eventually produce pre-piRNA. Once formed, pre-piRNA is translocated into the cytoplasm. In the cytoplasm, 5'-end of pre-piRNA binds to the piwi protein to form a piRNA-induced silencing complex (piRISC).15 Processed piRISC is transported back in to the nucleus through nuclear pores where it inhibits the transcription of transposon elements.16 Transposon elements have been identified to have a role in gene mutation leading to various diseases including cancers and infertility.17,18
Small Interfering RNAs (siRNAs)
Small interfering RNAs (siRNAs) are double-stranded, 21–25 nucleotide-long RNAs with two nucleotide overhangs at each hydroxylated 3'-end and phosphorylated 5'-end.19 Once in the cytoplasm, RNAse III dicer enzyme cleaves the long double-stranded RNA into siRNA. The siRNA is incorporated into RISC, which consists of Argonaute (Ago) protein, Dicer, and transactivating response RNA-binding protein (TRBP), leading to separation of double-stranded siRNA into the sense and antisense strand within the RISC complex. The antisense strand, with more stable 5'-end, forms the activated RISC complex, which in turn, targets mRNA through complementary base pairing.20–22
Small Nucleolar RNA (snoRNA)
Small nucleolar RNAs (snoRNAs) are 60–300 base-pair-long unique RNAs found only inside the nucleolus. There are two types of snoRNAs: (1) C/D box containing snoRNAs and (2) H/ACA box containing snoRNAs.23 Acting as a guide, snoRNAs direct selective chemical modification of nucleotides on other small housekeeping RNAs such as rRNAs. C/D box containing snoRNAs regulate sequence-specific 2'-O-methylation while H/ACA box snoRNAs regulate posttranscriptional isomerization of a uridine to a pseudouridine in rRNA.24
Circular RNAs (circRNAs)
Circular RNAs (circRNAs) are a large class of ncRNAs that originate from pre-mRNAs by a non-canonical splicing event called back-splicing. Consequently, loss of the terminal structures of a 5'-cap and a 3'-polyadenylation (poly-A) tail makes the circRNAs a covalently closed continuous ring structure.25 The unique configuration of circRNAs confers protection from exonuclease-mediated degradation and makes them remarkably stable molecules.4 Based on the sequence of origin, the circRNAs are categorized into exonic circRNAs (EcircRNA), intronic circRNAs (ciRNAs), exon-intronic circRNAs (ElciRNAs), intergenic circRNAs, and fusion circRNAs (f-cir-cRNAs). The EcircRNAs are the most abundant circRNAs predominantly located in the cytoplasm. The EcircRNAs function as miRNA sponge, modulate gene expression, regulate cell development and proliferation, as well as interact with RNA-binding proteins (RBPs).26 CiRNAs and ElciRNAs are predominantly present in the nucleus and regulate transcription and translation.27,28
Advances in genetic technologies and bioinformatics have shown that circRNAs may be generated from intergenic, intronic, coding regions, as well as untranslated regions of the DNA. The biosynthesis of circRNAs is explained by three proposed models based on splicing event orders: (a) lariat-driven circularization, also known as exon-skipping model; (b) intron interaction-driven circularization, also known as the direct back-splicing model; and (c) re-splicing-driven circularization.4,25 The biogenesis of circRNAs is illustrated in Fig. 2.
Figs 2A to E: The biogenesis of circRNAs. (A) Lariat-driven circularization also known as exon skipping. Exon skipping during canonical splicing forms lariats containing the skipped exons as well as mRNAs. The exon-containing lariats undergo back-splicing yielding EcircRNAs (intronic sequence removed) or ElciRNAs (intronic sequence retained); (B) Intron interaction-driven circularization. Direct base pairing between cis-acting; splicing regulatory elements (Alu repeats) or trans-acting factors (RBPs) couples flanking introns, followed by back-splicing and exon circularization. (C) Resplicing-driven circularization. Exons on mature RNA can undergo back-splicing and produce EcircRNA; (D) Biogenesis of ciRNA. The GU-rich (near 5, splice site, blue box) and the C-rich (near 3, splice site, red box) sequences can escape the debranching and degradation and form ciRNAs; (E) Biogenesis of intergenic circRNAs
Long Non-coding RNAs (lncRNAs)
lncRNAs affect many cellular processes at transcriptional, posttranscriptional, and translational levels. Most lncRNAs are located within intergenic stretches and are usually comprised of two-exon transcripts.29 These are interlaced, complex networks of overlapping sense and antisense transcripts that may also include protein-coding genes. Most ncRNAs, by definition, do not show protein-coding capacity, but some lncRNAs are now being identified to contain cryptic reading frames that may be translated into short, unstable micropeptides.30 Some sequence elements in lncRNAs may show conserved structure, but these do not show conserved functions. In other regions, some lncRNAs that are derived from syntenic regions and presumably have shared evolution, no longer show any similarity in sequences.31 These features suggest that many lncRNAs could possibly be non-functional or may have evolved from species-specific adaptive selection. The lncRNAs do seem to be important components of the address codes, which regulate directed trafficking, activation, and deactivation of protein complexes, genes, and chromosomes.32
Several types of lncRNAs have been identified. Based on proximity to the conventional protein-encoding mRNAs, lncRNAs can be classified as sense-, antisense-, or bidirectional lncRNAs.33 Sense lncRNA regions may overlap one or more exons of another coding transcript. In other instances, antisense lncRNAs can extend into coding genes. LncRNAs have also been classified by the genomic location as intronic- or long intervening/intergenic-ncRNAs (lincRNAs). Intronic lncRNAs are encoded in non-coding DNA sequences.34 The lincRNAs seem to be universal—these have been documented in plants, yeast, prokaryotes, and viruses, but the nucleotide sequences are not as well-conserved. Many lincRNAs are non-coding, autonomously transcribed long (>200 nucleotides) sequences that do not overlap with coding genes. Other classification group these into same-strand, isolated, convergent, or divergent categories, based on the location vis-à-vis the nearest protein-coding RNA.35 In terms of function, lincRNAs may regulate cellular processes such as the p53-mediated transcriptional responses to DNA damage.36
NCRNA ASSOCIATED WITH PREMATURITY THAT MAY INFLUENCE NEC
NEC is mainly a disease of premature neonates. Gestational age (GA) is inversely related to the incidence and severity of NEC. The intricate process of pregnancy maintenance and parturition necessitates a fine balance between many coordinated, consequential changes in hormones, tissue remodeling, metabolism, and immune system. Genetic factors, in conjugation with clinical and environmental variables, can alter the maternal-fetal interface and cause preterm birth. ncRNAs may play a regulatory role in gene expression controlling the process of pregnancy and birth. Since inflammation is a common theme for both, premature labor and NEC, various studies have evaluated miRNAs as well as lncRNAs that upregulate pro-inflammatory pathways and found that miRs-494, 142, 223, 15a, 329, 23a and lncRNAs-BF328678, BG258490, AA451649, BF667001, ENST00000423797, AX474492, BC107431, BX483760, DN918055, ENST00000437593 were associated with preterm labor (Luo 2013, Luo 2015).37–42 Similarly, chorioamnionitis increases the risk of NEC in premature infants due to deleterious effect of chronic inflammation on fetal cell programming.43 MiRNAs have been studied as modulators of chorioamnionitis, and consequently, its effects on fetal development and premature birth. Lee et al.44 examined autopsy samples of fetuses exposed to chorioamnionitis and noted increased expression of miR-223-3p in fetal thymus (2.55-fold), lung (1.93-fold), and liver (1.7-fold). This is an important finding as thymus plays a critical role in T cell development and aberrant T-helper cell response may cause inflammation.45,46 Montenegro et al.47 evaluated miRNA expression with advancing gestation and with chorioamnionitis in 39 pregnant women. Compared to controls, pregnant women with preterm labor and chorioamnionitis had increased expression of miR-223 (37-fold) and miR-338 (24-fold). In another study, 48 Korean pregnant women with chorioamnionitis and preterm birth had decreased expression of miR-548, but increased HMGB1 and inflammatory cytokines.48 These data suggest a need for further study of ncRNAs in the pathogenesis of premature birth and neonatal morbidities.
Maternal preeclampsia is an important cause of preterm birth. Qian et al. noted increased expression of hsa_circRNA_100782, hsa_circRNA_102682, and hsa_circRNA_104820 in human placental tissues from mothers with preeclampsia.49 Small for gestational age (SGA) neonates may be at increased risk of NEC.50,51 Wang et al.52 evaluated circRNAs in maternal and neonatal umbilical cord blood from SGA neonates and demonstrated that Hsa_circRNA15994-13, hsa_circ_0001359, and hsa_circ_0001360 were differentially expressed between SGA and AGA groups. The study also identified the target, hsa-miR-3619-5p, which plays an important role in the Wnt signaling pathway. These studies did not evaluate the neonatal outcomes, and future studies may be needed to examine neonatal outcomes.
NCRNAS ASSOCIATED WITH SPECIFIC HISTOPATHOLOGICAL FINDINGS SEEN IN NEC
NEC is characterized by exaggerated inflammation, coagulative necrosis, and hemorrhagic necrosis.1 In the following sections, ncRNAs that may be associated with the characteristic histopathological NEC features have been described.
ncRNAs Associated with Bowel Necrosis
The pathological process of NEC begins with intestinal epithelial cell (IEC) apoptosis, which results in mucosal defects and consequently, bacterial translocation from the gut lumen into the intestinal wall.53–59 This triggers an overwhelming inflammatory response and mucosal necrosis, and can ultimately lead to NEC.53,58,59 Apoptosis, programmed cell death, is immunologically a silent event, but NEC is characterized by exaggerated inflammation. To describe this unique pathoanatomical combination of NEC, a novel term, necroptosis, has been coined. Necroptosis may be caspase-independent in certain situations and can be triggered by death receptors such as transferrin-independent receptor-1 (NTFR1), interferon-production regulator (IFNR), Toll-like receptor (TLR) 3/4, Fas, and tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL). The ligation of death receptors activates the necrosome, a complex of the receptor-interacting serine/threonine kinases 1-3 (RIPK1-3) that in turn phosphorylates the mixed-linkage kinase domain-like protein (MLKL) to promote necroptosis.60,61
Werts et al.62 examined IEC necroptosis and noted TLR-induced activation of RIPK1, RIPK3, and MLKL, and the protective effect of human breast milk. Li et al.63 identified miR-141-3p as one of the agents that could possibly protect RIPK1 by downregulating RIPK1-MLKL-mediated necroptosis pathway. Chen et al.64 identified motor neuron and pancreas homeobox (MNX) 1, also known as HB9 or HLXB9, as another binding target of miR-141-3p, by showing that it suppresses the MNX1 gene. MiR-141-3p may also alleviate inflammation, apoptosis, and oxidative stress damage by regulating MNX1 expression. Wu et al.65 studied miR-431 in Chinese infants with stage 3 NEC (10 infants with NEC and an equal number of matched controls, and noted higher expression of miR-431 in the NEC group leading to suppressed forkhead box A1 (FOXA1) expression, and a significant effect downstream of miR-431-FOXA1 axis with exaggerated inflammation (increased expression of TNF, IL-6, IL-8, IL-10, NFKB2, and PLA2G2A), apoptosis (increased LGR5, decreased estrogen-related receptor gamma-ESRRG expression), and dysregulated tight junctions (decreased hepatocyte nuclear factor (HNF) 4A and PRKCZ expression). In another study, Ng et al.66 searched for novel NEC biomarkers. After studying 301 episodes (36 episodes of NEC, 265 episodes of non-NEC) in Chinese infants, they identified three potential early biomarkers, miR-1290, miR-1246, and miR-375. MiR-1290 was most accurate in the detection of NEC (sensitivity of 0.83, specificity of 0.92, PPV of 0.6, and NPV of 0.98 with a cutoff of >220 copies/μL). When they combined miR-1290 level of >650 copies/μL measured on day 0 and CRP level of >15.8 mg/L measured on day 1, they were able to correctly recognize 30/36 (83%) NEC cases. MiR-1290 has been studied in colorectal cancer and inflammatory bowel diseases (IBDs) and is noted to modulate inflammation, cell renewal, and apoptosis via FOXA1 pathway.67,68
ncRNAs Associated with Intestinal Inflammation in NEC
NEC is marked by an acute inflammatory response to microbial invasion. However, the determinants of the severity of inflammation are not fully understood.69 The pattern-recognition receptors (PRRs) are known to differentially recognize pathogens from other antigens and modulate the consequent immune responses. TLRs are one class of pattern-recognition receptors; TLR4 recognizes Gram-negative bacterial cell wall components such a lipopolysaccharide that may be involved in the pathogenesis of NEC. The activated TLRs recruit the myeloid differentiation (MD) factor and trigger downstream signaling to activate the nuclear factor-кB (NF-кB) and its related inflammatory responses.70 Therefore, the role of ncRNAs in the regulation of TLR-mediated pathways may be important in NEC pathogenesis.
The role of miR-124 on TLR-mediated inflammation and apoptosis via myosin phosphate target subunit 1 (MYPT1) and rho-associated coiled-coil-containing protein kinase 1 (ROCK1) was evaluated by Yin et al.71 using neonatal rat models of NEC. The study reported that miR-124 may protect against NEC by suppressing MYPT1, ROCK1, and TLR-9. Xu et al.72 evaluated the regulatory interactions of miRNAs and lncRNAs in NEC pathogenesis. They reported upregulation of the lncRNA MSTRG.42950 and MSTRG.104993 and downregulation of lncRNAs MSTRG.61378 and MSTRG.8198. There are recognizable binding patterns: lncRNA MSTRG.42950 with miR181a-5p; lncRNA MSTRG.104993 with miR-124-3p; and miR-194-5p with lncRNA. MSTRG.61378 may bind miR-362-3p, and lncRNA MSTRG.8198 binds miR-124-3p. These interactions likely modulate the TLR4 signaling pathway, TORC2 complex, notch signaling pathway, the p53 signaling pathway, and the mTOR pathway and, consequently, determine the severity of inflammation in NEC. More recently, Sun et al. studied the role of let-7d-5p/LGALS3/TLR4/NF-кB axis in the inflammatory cascades known to be active in NEC lesions. They noted decreased let-7d-5p and increased LGALS3 (galectin) in such lesions, possibly pointing to anti-inflammatory and protective roles of let-7d-5p.
TLR pathways also activate macrophages, and these cells, in turn, control gene expression and immune response modulation. Ng et al.73,74 evaluated the regulatory role of mcircRasGEF1B in the TLR4/LPS pathway. They identified increased mcircRASGEF1B in macrophages after LPS-induced activation. Depletion of mcircRasGEF1B dysregulated the TLR4/LPS pathway and caused macrophage dysfunction. Together, these findings provide future directions for large clinical studies with infants of different genetic backgrounds.
The nucleotide-binding oligomerization domain-containing (NOD) 2 is another cytosolic PRR that binds bacterial peptidoglycans and promotes pro-inflammatory cytokine production, inflammation, and innate immune defenses.75 MiRNAs also interact with the NOD2 pathway in adults with IBD. MiRNAs including miR-122, miR-192, miR-495, miR-671, miR-320, and miR-10a influence NOD2 expression, modulating inflammation and injury in IECs.76–81 Such studies are needed to evaluate the role of these miRNAs in NEC.
Mannose-binding lectin (MBL) is a circulating PRR that opsonizes pathogens and activates the lectin pathway of the complement system. It is an important regulator of inflammation, in a variety of conditions such as neonatal sepsis, pneumonia, NEC, and IBD.82–85 Prencipe et al. studied 107 neonates with NEC and showed that an SNP in the MBL2 gene increased serum levels of MBL in severe NEC.86 MiRNA regulation of MBL levels has been previously examined in hepatocellular carcinoma; miR-942-3p has been noted to bind MBL2.87 Studies are needed to evaluate how miRNAs may modulate the MBL pathway in NEC.
Pro-inflammatory cytokines are upregulated in NEC. The pathophysiological role of cytokines is unresolved; we still are unsure whether many of these cytokines are the cause, or the effect of inflammation in various conditions. Chen et al.64 showed that increasing the expression of miR-141-3p can reverse the overexpression of pro-inflammatory cytokines such as IL-1β, IL-6, and TNF-α in intestinal injury models. In another study, Wu et al.65 investigated miR-431 effects on TNF, IL-6, IL-8, and IL-10 and found it to increase IL-6 and TNF expression. Findings in these studies warrant future evaluation in vivo models and/or clinical studies in NEC.
ncRNAs Affecting Intestinal Microcirculation
Abnormalities in intestinal microcirculation due to maldevelopment or altered blood flow may contribute to NEC risk by causing intestinal ischemia and breach in mucosal integrity.88 Vascular endothelial growth factor-A (VEGFA) plays a key role in intestinal vasculature development.88,89 Association between decreased VEGF level and NEC has been established in both human as well as animal NEC models.90 NcRNAs may regulate VEGFA genes affecting intestinal vasculature development and vasoreactivity.
The association between miRNAs modulating VEGF and NEC has been investigated. Liu et al.91 reported downregulation of miR-429/200a/b and miR-141/200c clusters in four infants with NEC. The possible target genes for these two miRNA clusters, such as VEGFA, kinase insert domain receptor (KDR, also known as VEGFR2), FMS-related tyrosine kinase (FLT) 1, E-selectin (SELE), hepatocyte growth factor (HGF), were highly expressed in infants with NEC. Recently, these findings were confirmed by Zhao et al.92 and also showed the interaction of miR-200c-3p and miR-22a-3p with KDR genes. The study identified three additional potential targets in apoptotic pathway, tyrosine 3-monooxygenase (TH)/tryptophan 5-monooxygenase activation protein gamma (YWHAG), YWHA protein epsilon (YWHAE), and YWHA protein beta (YWHAB).
Hypoxia is a main angiogenesis stimulus causing VEGF-mediated angiogenesis in endothelial cells. Fiedler et al.93 identified two lncRNAs, LINC00323 and MIR503HG, in endothelial cells which are found to be highly sensitive to hypoxia and crucial for angiogenesis. Silencing of these two lncRNAs led to angiogenic defect, whereas endothelial cell treatment with VEGF increased their expression. Likewise, angiogenesis modulation by circRNAs has been explored in various pathological processes such as circ_100933, circ_100709, circ_104310 in infantile hemangioma;94 circ_0004158, circ_0005768, circ_0008737, circ_0005324, circ_0007799, circ_0005477, circ_0000668, circ_0012698, circ_0013414 in retinopathy of prematurity;95 circ_0005015, cZNF609, ZNF280c in diabetic retinopathy;96 ZNF609, ZNF292, HIPK3, circ_0010729, circ_0003575, circ_0054633, antisense noncoding RNA in the INK4 locus (ANRIL), CPWWP2A, circ_0068087, circ_0008360, circ_0000109, circ_0002317 in cardiovascular diseases;97 and SHKBP1, circ_002136 and SMARCA5 in tumorigenesis and metastasis.98 Similar studies are needed in NEC examining the regulatory role of ncRNAs in intestinal angiogenesis.
ncRNAs Associated with Intestinal Hemorrhages in NEC
Clinical features of severe NEC commonly include coagulopathy and thrombocytopenia. There exists a knowledge gap explaining pathophysiology of coagulopathy and thrombocytopenia in NEC. The only available evidence is from a study by Giuliani et al.99 who compared the expression of genes involved in coagulation in 11 infants with NEC with 22 controls and identified upregulation of hepatocyte growth factor (HGF), neutrophil-expressed elastase (ELANE), CD63, protein S (PROS1), and coagulation factor XII (F12) genes and downregulation of milk fat globule-EGF factor 8 (MFGE8), factor II (thrombin) receptor-like 1 (F2RL1), fibrinogen-like 2 (FGL2), plasminogen activator-tissue type (PLAT), protein C receptor (PROCR), serpin family D member 1 (SERPIND1), and hepatocyte nuclear factor-4a (HNF4A) genes. Out of these 12 genes, HNF4A is crucial for IEC maturation. Wu et al.65 showed that overexpression of miR-432 inhibits in the Caco-2 cell model of NEC. However, the study did not highlight any effect on coagulation cascade and thrombocytopenia.
There is evidence to suggest the involvement of miRNAs in thrombocytopenia other neonatal inflammatory disorders. Cui et al.100 identified a reduction in miR-130a expression in infants with sepsis who developed thrombocytopenia. MiR-130a targets IL-18 and/or IL-27 and was found to increase IL-18 expression without any change in IL-27 in the study. There has not been any study till date identifying specific ncRNAs associated with thrombocytopenia and coagulopathy in NEC providing an opportunity for future studies.
NCRNAS ASSOCIATED WITH GUT DYSBIOSIS
Gut microbiome is a unique, complex interdependent ecosystem. With more than 3 million genes, gut microbiome can shape the gene expression in the host and determine health and diseases.101 Dysbiosis is characterized by decreased diversity and overgrowth of pathogenic bacteria and has been linked to many inflammatory disorders, including NEC and IBD.102 The microbiome development is a dynamic process that begins even before birth and undergoes dramatic changes during infancy due to vast contribution from various factors such as gestational age, mode of delivery, type of feeding, and antibiotic exposure.103 Increasing information now associates genetics and the gut microbiome and vice versa.104–106 Liang et al. studied conventional, germ-free, and gnotobiotic mice to characterize lncRNAs that are regulated by gut microbiota and identified six upregulated and overlapped lncRNAs, n26353, n290292, n297037, n294754, n264146, and n288632. Interestingly, most of them were highly expressed in spleen and thymus, suggesting the role of microbiome in immune modulation via lncRNAs. Dempsey et al.107 demonstrated altered lncRNA expression in various organs such as the colon, liver, ileum, white fat tissue, jejunum, duodenum, and skeletal muscles. The mechanisms by which gut dysbiosis, lncRNA dysregulation, and intestinal inflammation may be linked need elucidation.
As mentioned earlier, miRNAs are the best-studied ncRNAs. These are more stable than other ncRNAs and are easier to measure in feces.108–110 However, the fecal miRNA levels may be affected by the fecal microbiome.108,111 The relationship between miRNA and gut dysbiosis has been studied in IBD and celiac disease. Mohan et al. linked intestinal dysbiosis and altered claudin-1 expression/epithelial junctions with increased inflammation-related miRNAs, miR-203, miR-204, miR-23a, and miR-29b.112 Studies in IBD have shown increased mir-144, mir-519, and mir-211, and downregulation of miR-577, miR-379-5p, miR-642-3p, and miR-26b-5p.113,114 Similar studies are needed to examine the role of miRNAs in gut dysbiosis and NEC.
NCRNAS ASSOCIATED WITH PROTECTIVE PROPERTIES OF HUMAN MILK
Human breast milk, a biological “elixir,” not only offers universally undisputed protection against NEC, but also reduces life-long health burden by preventing sudden infant death syndrome, bronchitis, lower respiratory tract infection, otitis media, atopy, and asthma.115 Human breast milk contains a large spectrum of miRNAs, either as free molecules or carried in exosomes or extracellular vesicles (EVs), and is known to shape the gut microbiome.116–120
The influence of miRNAs in milk-derived exosomes on intestinal maturation and inflammation has been studied in the setting of IBD and NEC. The therapeutic effects of milk-derived exosomes have been studied in murine models of colitis and reported higher expression of miR-375, let-7z, miR-148, and miR-320 in milk as well as milk-derived exosome treated colon while lower expression of miR-125b in colitis. These miRNAs lower the expression of IL-1β, IL-3, IL-6, IL-12, IL-15, and TNF.121–123 MiR-125b is known to regulate inflammation via NF-кB pathway that has a role in pathogenesis of NEC. MiR-148 also modulates immunity and has a role in metabolism and development. In intestinal cell culture models of NEC in rats and humans, milk-derived exosomes have shown a significant reduction in the incidence as well as the severity of NEC by anti-apoptotic, pro-proliferative, anti-inflammatory actions.124,125 Future studies exploring the miRNA content of exosomes and comparing formula and breast milk content will shed some light on this innovative therapeutic option for NEC.
Many researchers have examined other ncRNAs in breast milk. Karlsson et al.126 isolated 55 lncRNAs in EVs from human breast milk from 30 mothers within 2 months postpartum. These lncRNAs were present in more than 50% of the samples—CRNDE, DANCR, GAS5, HOTAIRM1, NCBP2-AS2, OIP5-AS1, PRKCQ-AS1, SNHG8, SRA1, TUG1, and ZFAS1. Later, Rubio et al.127 first discovered the presence of more than 1,000 small RNAs in breast milk including piRNAs, tRNAs, snoRNAs, and snRNAs with tRNAs being the most abundant. Recently, studies have also examined lncRNAs and circRNAs in bovine as well as porcine milk-derived exosomes.128,129 These data may be useful for future studies.
NCRNAS IN IBDS IN ADULTS
MiR-21 and miR-155 have been extensively studied in relation to IBD.130–134 MiR-21, located on chromosome 17q23.2 in humans, regulates inflammation in the innate immune system. It directly targets the p35 subunit of Th1-promoting IL-12 and NOS in intestinal endothelial cells by modulating P13K/Akt signaling pathway encoding mRNA.135 Increased miR-21 can alter the intestinal barrier and cause inflammation, oxidative stress, and cellular damage.131,135 Similarly, miR-155, located on chromosome 21q21.3 in humans, induces IL-17 secreting helper T cells maturation process via IL-23/17/6 axis and has been implicated in the pathogenesis of IBD.133,136–138 These findings are fascinating and provide future directions to confirm the role of miR-21 and miR-155 in neonates with NEC.
There is some information on the role of circRNAs and lncRNAs in IBD pathogenesis. Qiao et al.139 profiled circRNAs and their targeted miRNAs, genes, and pathways in 13 patients with Crohn’s disease (CD) and 13 controls; they found that hsa-circRNA-102685 may cause apoptosis via TLR and p53 signaling pathways via hsa-miR-146b-5p, hsa-miR-182-5p, and hsa-miR-146a-5p. Wang et al.140 identified hsa_circRNA_0007919 disrupting mucosal integrity via miR-138 and hsa_let-7a after comparing differential expression of circRNAs between inflamed and non-inflamed intestinal mucosa from 30 patients with ulcerative colitis. Yin et al.141 evaluated circRNAs in peripheral blood mononuclear cells obtained from IBD patients and discovered upregulation of hsa_circRNA_092520, hsa_circRNA_102610, hsa_circRNA_004662, and hsa_circRNA_103124, and correlation between circRNA_004662 and mTOR pathway via circRNA-miRNA-mRNA network prediction model. The mTOR plays a crucial role in the regulation of intestinal homeostasis and inflammation.142 Autophagy-related 16-like 1 (ATG16L1), one of the autophagy-related genes (ATGs), is essential for maintaining immune homeostasis and may confer protection against NEC.143,144 Genetic variation in ATG16L1 (Thr300Ala) increases risk of NEC, particularly in Caucasian infants.145 Using animal model of IBD, Li et al.146 showed that circRNA circPABPN1 blocked human antigen R (HuR) biding to atg16l1 mRNA and decreased ATG16L1 expression in the intestinal epithelium. Ye et al.147 identified circRNA_103516 as a potential biomarker after showing upregulation of circRNA_103516 in 180 patients with IBD and associated downregulation of miR-19b-a-5p.
Similarly, lncRNAs can be viewed as novel potential biomarkers for diagnosis as well as promising therapeutic targets for intestinal inflammatory conditions such as IBD and NEC. LncRNAs such as lncRNA NEAT1 (nuclear paraspeckle assembly transcript 1), lncRNA H19, and lncRNA SPRy4-IT1 are essential for intestinal epithelial regeneration and repair, thus maintaining intestinal epithelial barrier function.148 Studies have shown upregulation of lncRNA NEAT1 and lncRNA H19 in intestinal epithelium of IBD, whereas increased expression of lncRNA SPRy4-IT1 showed protective effect149–151 by modulating barrier function. Similarly, several differentially expressed lncRNAs were potentially associated with intestinal mucosal immune homeostasis, function of pro-inflammatory cytokines, and MHC protein complex.152 Specific inflammatory pathways affected by lncRNA dysregulation include those commonly identified in NEC pathogenesis such as NF-кB and TNF. Interleukin (IL)-1, IL-6, and IL-8 are often overexpressed in NEC and together with TNF; they stimulate NF-кB that leads to transcription of various inflammatory cytokines exacerbating the inflammation and tissue damage. Regulatory T lymphocytes (Tregs) are pivotal in keeping the excessive inflammation in check and maintenance of tolerance.45 Qiao et al.153 showed overexpression of lncRNA DQ786243 in 19 CD patients with active disease along with overexpression of cAMP response element binding protein (CREB) and forkhead box P3 (Foxp3), two key genes in function and development of Tregs, suggesting DQ786243, may be related to CD disease severity. Other lncRNA regulators of NF-кB have been implicated in the pathogenesis of IBD including lncRNA HIF1A-AS2, lncRNA ANRIL. Quan et al.154 demonstrated inactivation of NF-ĸB/JNK pathway by lncRNA HIF1A-AS2 leading to decreased expression of cytokines IL-1β, IL-6, IL-12, and TNF-α in mice colon samples. Qiao et al.155 demonstrated upregulation of lncRNA ANRIL in sigmoid colon mucosa obtained from 22 patients with UC and suggested that suppression of ANRIL may inhibit the development of UC by regulating miR-323-5p/TLR4/MyD88/ NF-ĸB pathway.
CONCLUSION
To expand our incomplete knowledge of complex NEC pathogenesis, we have reviewed the current literature on ncRNAs in NEC. The evidence remains imperfect due to scarcity of information on ncRNAs in NEC. Therefore, exploring the pathogenesis of other intestinal diseases such as IBD in adults and pediatric patients may provide a new direction to the future of NEC studies. Additionally, the complexity of NEC pathogenesis suggests that a single ncRNA may not explain NEC entirely. The evolution of high-throughput, in-depth next-generation sequencing techniques and bioinformatics may elucidate the interactions between different ncRNAs and their molecular mechanism in the pathogenesis of NEC.
REFERENCES
1. Denning PW, Maheshwari A. Necrotizing enterocolitis: hope on the horizon. Clin Perinatol 2013;40(1):xvii–xix. DOI: 10.1016/j.clp.2013.01.001.
2. Esteller M. Non-coding RNAs in human disease. Nat Rev Genet 2011;12(12):861–874. DOI: 10.1038/nrg3074.
3. Boivin V, Faucher-Giguère L, Scott M, et al. The cellular landscape of mid‐size noncoding RNA. WIREs RNA 2019;10(4):e1530. DOI: 10.1002/wrna.1530.
4. Jeck WR, Sorrentino JA, Wang K, et al. Circular RNAs are abundant, conserved, and associated with ALU repeats. RNA 2013;19(2):141–157. DOI: 10.1261/rna.035667.112.
5. Feng J, Naiman DQ, Cooper B. Coding DNA repeated throughout intergenic regions of the Arabidopsis thaliana genome: evolutionary footprints of RNA silencing. Mol Biosyst 2009;5(12):1679–1687. DOI: 10.1039/b903031j.
6. Jonas S, Izaurralde E. Towards a molecular understanding of microRNA-mediated gene silencing. Nat Rev Genet 2015;16(7):421–433. DOI: 10.1038/nrg3965.
7. Ozsolak F, Poling LL, Wang Z, et al. Chromatin structure analyses identify miRNA promoters. Genes Dev 2008;22(22):3172–3183. DOI: 10.1101/gad.1706508.
8. Yi R, Qin Y, Macara IG, et al. Exportin-5 mediates the nuclear export of pre-microRNAs and short hairpin RNAs. Genes Dev 2003;17(24):3011–3016. DOI: 10.1101/gad.1158803.
9. Gregory RI, Chendrimada TP, Cooch N, et al. Human RISC couples microRNA biogenesis and posttranscriptional gene silencing. Cell 2005;123(4):631–640. DOI: 10.1016/j.cell.2005.10.022.
10. Denli AM, Tops BB, Plasterk RH, et al. Processing of primary microRNAs by the Microprocessor complex. Nature 2004;432(7014):231–235. DOI: 10.1038/nature03049.
11. Peterson SM, Thompson JA, Ufkin ML, et al. Common features of microRNA target prediction tools. Front Genet 2014;5:23. DOI: 10.3389/fgene.2014.00023.
12. Lewis BP, Shih IH, Jones-Rhoades MW, et al. Prediction of mammalian microRNA targets. Cell 2003;115(7):787–798. DOI: 10.1016/s0092-8674(03)01018-3.
13. Friedman RC, Farh KKH, Burge CB, et al. Most mammalian mRNAs are conserved targets of microRNAs. Genome Res 2009;19(1):92–105. DOI: 10.1101/gr.082701.108.
14. Kondkar AA, Abu-Amero KK. Utility of circulating microRNAs as clinical biomarkers for cardiovascular diseases. Biomed Res Int 2015;2015:821823. DOI: 10.1155/2015/821823.
15. Guo B, Li D, Du L, et al. piRNAs: biogenesis and their potential roles in cancer. Cancer Metastasis Rev 2020;39(2):567–575. DOI: 10.1007/s10555-020-09863-0.
16. Hirakata S, Ishizu H, Fujita A, et al. Requirements for multivalent Yb body assembly in transposon silencing in Drosophila. EMBO reports 2019;20(7):e47708. DOI: 10.15252/embr.201947708.
17. Théron E, Maupetit-Mehouas S, Pouchin P, et al. The interplay between the Argonaute proteins Piwi and Aub within Drosophila germarium is critical for oogenesis, piRNA biogenesis and TE silencing. Nucleic Acids Res 2018;46(19):10052–10065. DOI: 10.1093/nar/gky695.
18. Weng W, Li H, Goel A. Piwi-interacting RNAs (piRNAs) and cancer: emerging biological concepts and potential clinical implications. Biochim Biophys Acta (BBA) – Rev Cancer 2019;1871(1):160–169. DOI: 10.1016/j.bbcan.2018.12.005.
19. Hombach S, Kretz M. Non-coding RNAs: classification, biology and functioning. Springer International Publishing; 2016. p. 3–17.
20. Shen J, Zhang W, Qi R, et al. Engineering functional inorganic–organic hybrid systems: advances in siRNA therapeutics. Chem Soc Rev 2018;47(6):1969–1995. DOI: 10.1039/c7cs00479f.
21. Nikam RR, Gore KR. Journey of siRNA: clinical developments and targeted delivery. Nucleic Acid Ther 2018;28(4):209–224. DOI: 10.1089/nat.2017.0715.
22. Saw PE, Song EW. siRNA therapeutics: a clinical reality. Sci China Life Sci 2020;63(4):485–500. DOI: 10.1007/s11427-018-9438-y.
23. Ramakrishnan S. Sno(RNA)wing and pancreatic cancer metastasis. Gastroenterology 2017;153(1):12–14. DOI: 10.1053/j.gastro.2017.05.039.
24. Bratkovič T, Rogelj B. The many faces of small nucleolar RNAs. Biochim Biophys Acta (BBA) 2014;1839(6):438–443. DOI: 10.1016/j.bbagrm.2014.04.009.
25. Kristensen LS, Andersen MS, Stagsted LVW, et al. The biogenesis, biology and characterization of circular RNAs. Nat Rev Genet 2019;20(11):675–691. DOI: 10.1038/s41576-019-0158-7.
26. Chen I, Chen CY, Chuang TJ. Biogenesis, identification, and function of exonic circular RNAs. Wiley Interdiscip Rev RNA 2015;6(5):563–579. DOI: 10.1002/wrna.1294.
27. Bolha L, Ravnik-Glavač M, Glavač D. Circular RNAs: biogenesis, function, and a role as possible cancer biomarkers. Int J Genomics 2017;2017:1–19. DOI: 10.1155/2017/6218353.
28. Li Z, Huang C, Bao C, et al. Exon-intron circular RNAs regulate transcription in the nucleus. Nat Struct Mol Biol 2015;22(3):256–264. DOI: 10.1038/nsmb.2959.
29. Kapranov P, Cheng J, Dike S, et al. RNA maps reveal new RNA classes and a possible function for pervasive transcription. Science 2007;316(5830):1484–1488. DOI: 10.1126/science.1138341.
30. Jabandziev P, Bohosova J, Pinkasova T, et al. The emerging role of noncoding RNAs in pediatric inflammatory bowel disease. Inflamm Bowel Dis 2020;26(7):985–993. DOI: 10.1093/ibd/izaa009.
31. Ulitsky I. Evolution to the rescue: using comparative genomics to understand long non-coding RNAs. Nat Rev Genet 2016;17(10):601–614. DOI: 10.1038/nrg.2016.85.
32. Batista PJ, Chang HY. Long noncoding RNAs: cellular address codes in development and disease. Cell 2013;152(6):1298–1307. DOI: 10.1016/j.cell.2013.02.012.
33. Yarani R, Mirza AH, Kaur S, et al. The emerging role of lncRNAs in inflammatory bowel disease. Exp Mol Med 2018;50(12):1–14. DOI: 10.1038/s12276-018-0188-9.
34. Beermann J, Piccoli MT, Viereck J, et al. Non-coding RNAs in development and disease: background, mechanisms, and therapeutic approaches. Physiol Rev 2016;96(4):1297–1325. DOI: 10.1152/physrev.00041.2015.
35. Ransohoff JD, Wei Y, Khavari PA. The functions and unique features of long intergenic non-coding RNA. Nat Rev Mol Cell Biol 2018;19(3):143–157. DOI: 10.1038/nrm.2017.104.
36. Huarte M, Guttman M, Feldser D, et al. A large intergenic noncoding RNA induced by p53 mediates global gene repression in the p53 response. Cell 2010;142(3):409–419. DOI: 10.1016/j.cell.2010.06.040.
37. Sanders AP, Gennings C, Svensson K, et al. Bacterial cytokine mixtures predict the length of gestation and are associated with miRNA expression in the cervix. Epigenomics 2017;9(1):33–45. DOI: 10.2217/epi-2016-0095.
38. Elovitz MA, Anton L, Bastek J, et al. Can microRNA profiling in maternal blood identify women at risk for preterm birth? Am J Obstet Gynecol 2015;212(6):782.e1–782.e5. DOI: 10.1016/j.ajog.2015.01.023.
39. Haneklaus M, Gerlic M, O’Neill LA, et al. miR-223: infection, inflammation and cancer. J Intern Med 2013;274(3):215–226. DOI: 10.1111/joim.12099.
40. Garg M, Potter JA, Abrahams VM. Identification of microRNAs that regulate TLR2-mediated trophoblast apoptosis and inhibition of IL-6 mRNA. PLoS One 2013;8(10):e77249. DOI: 10.1371/journal.pone.0077249.
41. Mayor-Lynn K, Toloubeydokhti T, Cruz AC, et al. Expression profile of MicroRNAs and mRNAs in human placentas from pregnancies complicated by preeclampsia and preterm labor. Reprod Sci 2011;18(1):46–56. DOI: 10.1177/1933719110374115.
42. Renthal NE, Chen CC, Williams KC, et al. miR-200 family and targets, ZEB1 and ZEB2, modulate uterine quiescence and contractility during pregnancy and labor. Proc Natl Acad Sci 2010;107(48):20828–20833. DOI: 10.1073/pnas.1008301107.
43. Been JV, Lievense S, Zimmermann LJ, et al. Chorioamnionitis as a risk factor for necrotizing enterocolitis: a systematic review and meta-analysis. J Pediatr 2013;162(2):236–242.e2. DOI: 10.1016/j.jpeds.2012.07.012.
44. Lee J, Kim CJ, Kim JS, et al. Increased miR-223 expression in foetal organs is a signature of acute chorioamnionitis with systemic consequences. J Cell Mol Med 2017;22(2):1179–1189. DOI: 10.1111/jcmm.13377.
45. Pang Y, Du X, Xu X, et al. Impairment of regulatory T cells in patients with neonatal necrotizing enterocolitis. Int Immunopharmacol 2018;63:19–25. DOI: 10.1016/j.intimp.2018.07.029.
46. Ma F, Li S, Gao X, et al. Interleukin-6-mediated CCR9(+) interleukin-17-producing regulatory T cells polarization increases the severity of necrotizing enterocolitis. EBioMedicine 2019;44:71–85. DOI: 10.1016/j.ebiom.2019.05.042.
47. Montenegro D, Romero R, Pineles BL, et al. Differential expression of microRNAs with progression of gestation and inflammation in the human chorioamniotic membranes. Am J Obstet Gynecol 2007;197(3):289.e1–289.e6. DOI: 10.1016/j.ajog.2007.06.027.
48. Son GH, Kim Y, Lee JJ, et al. MicroRNA-548 regulates high mobility group box 1 expression in patients with preterm birth and chorioamnionitis. Sci Rep 2019;9(1):19746. DOI: 10.1038/s41598-019-56327-9.
49. Qian Y, Lu Y, Rui C, et al. Potential significance of circular RNA in human placental tissue for patients with preeclampsia. Cell Physiol Biochem 2016;39(4):1380–1390. DOI: 10.1159/000447842.
50. Samuels N, van de Graaf RA, de Jonge RCJ, et al. Risk factors for necrotizing enterocolitis in neonates: a systematic review of prognostic studies. BMC Pediatr 2017;17(1):105. DOI: 10.1186/s12887-017-0847-3.
51. Boghossian NS, Geraci M, Edwards EM, et al. Morbidity and mortality in small for gestational age infants at 22 to 29 weeks’ gestation. Pediatrics 2018;141(2):e20172533. DOI: 10.1542/peds.2017-2533.
52. Wang Y, Li SF, Dang YJ, et al. Differentially expressed circular RNAs in maternal and neonatal umbilical cord plasma from SGA compared with AGA. J Cell Biochem 2020;121(1):713–722. DOI: 10.1002/jcb.29317.
53. MohanKumar K, Namachivayam K, Cheng F, et al. Trinitrobenzene sulfonic acid-induced intestinal injury in neonatal mice activates transcriptional networks similar to those seen in human necrotizing enterocolitis. Pediatr Res 2016;81(1):99–112. DOI: 10.1038/pr.2016.189.
54. MohanKumar K, Namachivayam K, Song T, et al. A murine neonatal model of necrotizing enterocolitis caused by anemia and red blood cell transfusions. Nat Commun 2019;10(1):3494. DOI: 10.1038/s41467-019-11199-5.
55. De Plaen IG, Liu SX, Tian R, et al. Inhibition of nuclear factor-kappaB ameliorates bowel injury and prolongs survival in a neonatal rat model of necrotizing enterocolitis. Pediatr Res 2007;61(6):716–721. DOI: 10.1203/pdr.0b013e3180534219.
56. Jilling T, Lu J, Jackson M, et al. Intestinal epithelial apoptosis initiates gross bowel necrosis in an experimental rat model of neonatal necrotizing enterocolitis. Pediatr Res 2004;55(4):622–629. DOI: 10.1203/01.PDR.0000113463.70435.74.
57. Jilling T, Simon D, Lu J, et al. The roles of bacteria and TLR4 in rat and murine models of necrotizing enterocolitis. J Immunol 2006;177(5):3273–3282. DOI: 10.4049/jimmunol.177.5.3273.
58. MohanKumar K, Namachivayam K, Chapalamadugu KC, et al. Smad7 interrupts TGF-β signaling in intestinal macrophages and promotes inflammatory activation of these cells during necrotizing enterocolitis. Pediatr Res 2016;79(6):951–961. DOI: 10.1038/pr.2016.18.
59. Namachivayam K, Blanco CL, MohanKumar K, et al. Smad7 inhibits autocrine expression of TGF-beta2 in intestinal epithelial cells in baboon necrotizing enterocolitis. Am J Physiol Gastrointest Liver Physiol 2013;304(2):G167–G180. DOI: 10.1152/ajpgi.00141.2012.
60. Frank D, Vince JE. Pyroptosis versus necroptosis: similarities, differences, and crosstalk. Cell Death Differ 2019;26(1):99–114. DOI: 10.1038/s41418-018-0212-6.
61. Dhuriya YK, Sharma D. Necroptosis: a regulated inflammatory mode of cell death. J Neuroinflammation 2018;15(1):199. DOI: 10.1186/s12974-018-1235-0.
62. Werts AD, Fulton WB, Ladd MR, et al. A novel role for necroptosis in the pathogenesis of necrotizing enterocolitis. Cell Mol Gastroenterol Hepatol 2020;9(3):403–423. DOI: 10.1016/j.jcmgh.2019.11.002.
63. Li X, Wang Y, Wang Y, et al. MiR-141-3p ameliorates RIPK1-mediated necroptosis of intestinal epithelial cells in necrotizing enterocolitis. Aging (Albany NY) 2020;12(18):18073–18083. DOI: 10.18632/aging.103608.
64. Chen H, Zeng L, Zheng W, et al. Increased expression of microRNA-141-3p improves necrotizing enterocolitis of neonates through targeting MNX1. Front Pediatr 2020;8:385. DOI: 10.3389/fped.2020.00385.
65. Wu YZ, Chan KYY, Leung KT, et al. Dysregulation of miR-431 and target gene FOXA1 in intestinal tissues of infants with necrotizing enterocolitis. Federation Am Soc Exp Biol J2019;33(4):5143–5152. DOI: 10.1096/fj.201801470R.
66. Ng PC, Chan KYY, Yuen TP, et al. Plasma miR-1290 is a novel and specific biomarker for early diagnosis of necrotizing enterocolitis-biomarker discovery with prospective cohort evaluation. J Pediatr 2019;205:83–90.e10. DOI: 10.1016/j.jpeds.2018.09.031.
67. Imaoka H, Toiyama Y, Fujikawa H, et al. Circulating microRNA-1290 as a novel diagnostic and prognostic biomarker in human colorectal cancer. Ann Oncol 2016;27(10):1879–1886. DOI: 10.1093/annonc/mdw279.
68. van der Sluis M, Vincent A, Bouma J, et al. Forkhead box transcription factors Foxa1 and Foxa2 are important regulators of Muc2 mucin expression in intestinal epithelial cells. Biochem Biophys Res Commun 2008;369(4):1108–1113. DOI: 10.1016/j.bbrc.2008.02.158.
69. Maheshwari A, Kelly DR, Nicola T, et al. TGF-β2 suppresses macrophage cytokine production and mucosal inflammatory responses in the developing intestine. Gastroenterology 2011;140(1):242–253. DOI: 10.1053/j.gastro.2010.09.043.
70. Mara MA, Good M, Weitkamp JH. Innate and adaptive immunity in necrotizing enterocolitis. Semin Fetal Neonatal Med 2018;23(6):394–399. DOI: 10.1016/j.siny.2018.08.002.
71. Yin Y, Qin Z, Xu X, et al. Inhibition of miR-124 improves neonatal necrotizing enterocolitis via an MYPT1 and TLR9 signal regulation mechanism. J Cell Physiol 2019;234(7):10218–10224. DOI: 10.1002/jcp.27691.
72. Xu Y, Liu Y, Xie H, et al. Profile analysis reveals endogenous RNAs regulate necrotizing enterocolitis progression. Biomed Pharmacother 2020;125:109975. DOI: 10.1016/j.biopha.2020.109975.
73. Ng WL, Marinov GK, Chin YM, et al. Transcriptomic analysis of the role of RasGEF1B circular RNA in the TLR4/LPS pathway. Sci Rep 2017;7(1):12227. DOI: 10.1038/s41598-017-12550-w.
74. Ng WL, Marinov GK, Liau ES, et al. Inducible RasGEF1B circular RNA is a positive regulator of ICAM-1 in the TLR4/LPS pathway. RNA Biol 2016;13(9):861–871. DOI: 10.1080/15476286.2016.1207036.
75. Takeuchi O, Akira S. Pattern recognition receptors and inflammation. Cell 2010;140(6):805–820. DOI: 10.1016/j.cell.2010.01.022.
76. Chuang AY, Chuang JC, Zhai Z, et al. NOD2 expression is regulated by microRNAs in colonic epithelial HCT116 cells. Inflamm Bowel Dis 2014;20(1):126–135. DOI: 10.1097/01.MIB.0000436954.70596.9b.
77. Wu W, He C, Liu C, et al. miR-10a inhibits dendritic cell activation and Th1/Th17 cell immune responses in IBD. Gut 2015;64(11):1755–1764. DOI: 10.1136/gutjnl-2014-307980.
78. Xu X, Ma C, Liu C, et al. Knockdown of long noncoding RNA XIST alleviates oxidative low-density lipoprotein-mediated endothelial cells injury through modulation of miR-320/NOD2 axis. Biochem Biophys Res Commun 2018;503(2):586–592. DOI: 10.1016/j.bbrc.2018.06.042.
79. Ghorpade DS, Sinha AY, Holla S, et al. NOD2-nitric oxide-responsive microRNA-146a activates sonic Hedgehog signaling to orchestrate inflammatory responses in murine model of inflammatory bowel disease. J Biol Chem 2013;288(46):33037–33048. DOI: 10.1074/jbc.M113.492496.
80. Chen Y, Wang C, Liu Y, et al. miR-122 targets NOD2 to decrease intestinal epithelial cell injury in Crohn’s disease. Biochem Biophys Res Commun 2013;438(1):133–139. DOI: 10.1016/j.bbrc.2013.07.040.
81. Brain O, Owens BM, Pichulik T, et al. The intracellular sensor NOD2 induces MicroRNA-29 expression in human dendritic cells to limit IL-23 release. Immunity 2013;39(3):521–536. DOI: 10.1016/j.immuni.2013.08.035.
82. Frakking FN, Brouwer N, Zweers D, et al. High prevalence of mannose-binding lectin (MBL) deficiency in premature neonates. Clin Exp Immunol 2006;145(1):5–12. DOI: 10.1111/j.1365-2249.2006.03093.x.
83. de Benedetti F, Auriti C, D’Urbano LE, et al. Low serum levels of mannose binding lectin are a risk factor for neonatal sepsis. Pediatr Res 2007;61(3):325–328. DOI: 10.1203/pdr.0b013e318030d12f.
84. Schlapbach LJ, Latzin P, Regamey N, et al. Mannose-binding lectin cord blood levels and respiratory symptoms during infancy: a prospective birth cohort study. Pediatr Allergy Immunol 2009;20(3):219–226. DOI: 10.1111/j.1399-3038.2008.00782.x.
85. Takahashi K. Mannose-binding lectin and the balance between immune protection and complication. Expert Rev Anti Infect Ther 2011;9(12):1179–1190. DOI: 10.1586/eri.11.136.
86. Prencipe G, Azzari C, Moriondo M, et al. Association between mannose-binding lectin gene polymorphisms and necrotizing enterocolitis in preterm infants. J Pediatr Gastroenterol Nutr 2012;55(2):160–165. DOI: 10.1097/MPG.0b013e31824e5f7a.
87. Xu CY, Dong JF, Chen ZQ, et al. MiR-942-3p promotes the proliferation and invasion of hepatocellular carcinoma cells by targeting MBL2. Cancer Control 2019;26(1):107327481984659. DOI: 10.1177/1073274819846593.
88. Bowker RM, Yan X, De Plaen IG. Intestinal microcirculation and necrotizing enterocolitis: the vascular endothelial growth factor system. Semin Fetal Neonatal Med 2018;23(6):411–415. DOI: 10.1016/j.siny.2018.08.008.
89. Crafts TD, Jensen AR, Blocher-Smith EC, et al. Vascular endothelial growth factor: therapeutic possibilities and challenges for the treatment of ischemia. Cytokine 2015;71(2):385–393. DOI: 10.1016/j.cyto.2014.08.005.
90. Sabnis A, Carrasco R, Liu SX, et al. Intestinal vascular endothelial growth factor is decreased in necrotizing enterocolitis. Neonatology 2015;107(3):191–198. DOI: 10.1159/000368879.
91. Liu H, Wang YB. Systematic large-scale meta-analysis identifies miRNA-429/200a/b and miRNA-141/200c clusters as biomarkers for necrotizing enterocolitis in newborn. Biosci Rep 2019;39(9):BSR20191503. DOI: 10.1042/BSR20191503.
92. Zhao J, Yin L, He L. The microRNA landscapes profiling reveals potential signatures of necrotizing enterocolitis in infants. J Comput Biol 2020;27(1):30–39. DOI: 10.1089/cmb.2019.0183.
93. Fiedler J, Breckwoldt K, Remmele CW, et al. Development of long noncoding RNA-based strategies to modulate tissue vascularization. J Am Coll Cardiol 2015;66(18):2005–2015. DOI: 10.1016/j.jacc.2015.07.081.
94. Fu C, Lv R, Xu G, et al. Circular RNA profile of infantile hemangioma by microarray analysis. PLoS One 2017;12(11):e0187581. DOI: 10.1371/journal.pone.0187581.
95. Zhou H, Song H, Wu Y, et al. Oxygen-induced circRNA profiles and coregulatory networks in a retinopathy of prematurity mouse model. Exp Ther Med 2019;18(3):2037–2050. DOI: 10.3892/etm.2019.7819.
96. Guo N, Liu XF, Pant OP, et al. Circular RNAs: novel promising biomarkers in ocular diseases. Int J Med Sci 2019;16(4):513–518. DOI: 10.7150/ijms.29750.
97. Zhang TR, Huang WQ. Angiogenic circular RNAs: a new landscape in cardiovascular diseases. Microvasc Res 2020;129:103983. DOI: 10.1016/j.mvr.2020.103983.
98. Ma Z, Shuai Y, Gao X, et al. Circular RNAs in the tumour microenvironment. Mol Cancer 2020;19(1):8. DOI: 10.1186/s12943-019-1113-0.
99. Giuliani S, Tan YW, Zheng D, et al. Coagulation gene expression profiling in infants with necrotizing enterocolitis. J Pediatr Gastroenterol Nutr 2016;63(6):e169–e175. DOI: 10.1097/MPG.0000000000001215.
100. Cui YL, Wang B, Gao HM, et al. Interleukin-18 and miR-130a in severe sepsis patients with thrombocytopenia. Patient Prefer Adherence 2016(10):313–319. DOI: 10.2147/PPA.S95588.
101. Celiker C, Kalkan R. Genetic and epigenetic perspective of microbiota. Appl Microbiol Biotechnol 2020;104(19):8221–8229. DOI: 10.1007/s00253-020-10849-9.
102. Ray K. IBD. Understanding gut microbiota in new-onset Crohn’s disease. Nat Rev Gastroenterol Hepatol 2014;11(5):268. DOI: 10.1038/nrgastro.2014.45.
103. Ursell LK, Metcalf JL, Parfrey LW, et al. Defining the human microbiome. Nutr Rev 2012;70(Suppl. 1):S38–S44. DOI: 10.1111/j.1753-4887.2012.00493.x.
104. Goodrich JK, Davenport ER, Beaumont M, et al. Genetic determinants of the gut microbiome in UK twins. Cell Host Microbe 2016;19(5):731–743. DOI: 10.1016/j.chom.2016.04.017.
105. Goodrich JK, Waters JL, Poole AC, et al. Human genetics shape the gut microbiome. Cell 2014;159(4):789–799. DOI: 10.1016/j.cell.2014.09.053.
106. Turpin W, Espin-Garcia O, Xu W, et al. Association of host genome with intestinal microbial composition in a large healthy cohort. Nat Genet 2016;48(11):1413–1417. DOI: 10.1038/ng.3693.
107. Dempsey J, Zhang A, Cui JY. Coordinate regulation of long non-coding RNAs and protein-coding genes in germ-free mice. BMC Genom 2018;19(1):834. DOI: 10.1186/s12864-018-5235-3.
108. Liu S, Weiner HL. Control of the gut microbiome by fecal microRNA. Microbial Cell 2016;3(4):176–177. DOI: 10.15698/mic2016.04.492.
109. Link A, Becker V, Goel A, et al. Feasibility of fecal MicroRNAs as novel biomarkers for pancreatic cancer. PLoS One 2012;7(8):e42933. DOI: 10.1371/journal.pone.0042933.
110. Ahmed F, Jeffries CD, Vos PW, et al. Diagnostic MicroRNA markers for screening sporadic humancolon cancer and active ulcerative colitis in stool and tissue. Cancer Genom Proteom 2009;6(5):281–295. PMID: 19996134.
111. Liu S, da Cunha AP, Rezende RM, et al. The host shapes the gut microbiota via fecal MicroRNA. Cell Host Microbe 2016;19(1):32–43. DOI: 10.1016/j.chom.2015.12.005.
112. Mohan M, Chow CT, Ryan CN, et al. Dietary gluten-induced gut dysbiosis is accompanied by selective upregulation of microRNAs with intestinal tight junction and bacteria-binding motifs in rhesus macaque model of celiac disease. Nutrients 2016;8(11):684. DOI: 10.3390/nu8110684.
113. Rojas-Feria M, Romero-García T, Fernández Caballero-Rico JÁ, et al. Modulation of faecal metagenome in Crohn’s disease: role of microRNAs as biomarkers. World J Gastroenterol 2018;24(46):5223–5233. DOI: 10.3748/wjg.v24.i46.5223.
114. Ambrozkiewicz F, Karczmarski J, Kulecka M, et al. In search for interplay between stool microRNAs, microbiota and short chain fatty acids in Crohn’s disease – a preliminary study. BMC Gastroenterol 2020;20(1):307. DOI: 10.1186/s12876-020-01444-3.
115. Galley JD, Besner GE. The therapeutic potential of breast milk-derived extracellular vesicles. Nutrients 2020;12(3):745. DOI: 10.3390/nu12030745.
116. Le Doare K, Holder B, Bassett A, et al. Mother’s milk: a purposeful contribution to the development of the infant microbiota and immunity. Front Immunol 2018;9:361. DOI: 10.3389/fimmu.2018.00361.
117. Carrillo-Lozano E, Sebastián-Valles F, Knott-Torcal C. Circulating microRNAs in breast milk and their potential impact on the infant. Nutrients 2020;12(10):3066. DOI: 10.3390/nu12103066.
118. Tomé-Carneiro J, Fernández-Alonso N, Tomás-Zapico C, et al. Breast milk microRNAs harsh journey towards potential effects in infant development and maturation. Lipid encapsulation can help. Pharmacol Res 2018;132:21–32. DOI: 10.1016/j.phrs.2018.04.003.
119. Wu F, Zhi X, Xu R, et al. Exploration of microRNA profiles in human colostrum. Ann Transl Med 2020;8(18):1170–1170. DOI: 10.21037/atm-20-5709.
120. Liao Y,Du X, Li J, et al. Human milk exosomes and their microRNAs survive digestion in vitro and are taken up by human intestinal cells. Mol Nutr Food Res 2017;61(11):1700082. DOI: 10.1002/mnfr.201700082.
121. Reif S, Elbaum-Shiff Y, Koroukhov N, et al. Cow and human milk-derived exosomes ameliorate colitis in DSS murine model. Nutrients 2020;12(9):2589. DOI: 10.3390/nu12092589.
122. Benmoussa A, Diallo I, Salem M, et al. Concentrates of two subsets of extracellular vesicles from cow’s milk modulate symptoms and inflammation in experimental colitis. Sci Rep 2019;9(1):14661. DOI: 10.1038/s41598-019-51092-1.
123. Stremmel W, Weiskirchen R, Melnik BC. Milk exosomes prevent intestinal inflammation in a genetic mouse model of ulcerative colitis: a pilot experiment. Inflamm Intest Dis 2020;5(3):117–123. DOI: 10.1159/000507626.
124. Pisano C, Galley J, Elbahrawy M, et al. Human breast milk-derived extracellular vesicles in the protection against experimental necrotizing enterocolitis. J Pediatr Surg 2020;55(1):54–58. DOI: 10.1016/j.jpedsurg.2019.09.052.
125. Li B, Hock A, Wu RY, et al. Bovine milk-derived exosomes enhance goblet cell activity and prevent the development of experimental necrotizing enterocolitis. PLoS One 2019;14(1):e0211431. DOI: 10.1371/journal.pone.0211431.
126. Karlsson O, Rodosthenous RS, Jara C, et al. Detection of long non-coding RNAs in human breastmilk extracellular vesicles: implications for early child development. Epigenetics 2016;11(10):721–729. DOI: 10.1080/15592294.2016.1216285.
127. Rubio M, Bustamante M, Hernandez-Ferrer C, et al. Circulating miRNAs, isomiRs and small RNA clusters in human plasma and breast milk. PLoS One 2018;13(3):e0193527. DOI: 10.1371/journal.pone.0193527.
128. Wang Y, Wei LD, Huan WY, et al. The landscape of circular RNAs and mRNAs in bovine milk exosomes. J Food Compos Analysis 2019;76:33–38. DOI: 10.1016/j.jfca.2018.12.004.
129. Zeng B, Chen T, Luo J, et al. Exploration of long non-coding RNAs and circular RNAs in porcine milk exosomes. Front Genet 2020;11:652. DOI: 10.3389/fgene.2020.00652.
130. James JP, Riis LB, Malham M, et al. MicroRNA biomarkers in IBD—differential diagnosis and prediction of colitis-associated cancer. Int J Mol Sci 2020;21(21):7893. DOI: 10.3390/ijms21217893.
131. Thorlacius-Ussing G, Schnack Nielsen B, Andersen V, et al. Expression and localization of miR-21 and miR-126 in mucosal tissue from patients with inflammatory bowel disease. Inflamm Bowel Dis 2017;23(5):739–752. DOI: 10.1097/MIB.0000000000001086.
132. Béres NJ, Szabó D, Kocsis D, et al. Role of altered expression of miR-146a, miR-155, and miR-122 in pediatric patients with inflammatory bowel disease. Inflamm Bowel Dis 2016;22(2):327–335. DOI: 10.1097/MIB.0000000000000687.
133. Valmiki S, Ahuja V, Paul J. MicroRNA exhibit altered expression in the inflamed colonic mucosa of ulcerative colitis patients. World J Gastroenterol 2017;23(29):5324. DOI: 10.3748/wjg.v23.i29.5324.
134. Schönauen K, Le N, von Arnim U, et al. Circulating and fecal microRNAs as biomarkers for inflammatory bowel diseases. Inflamm Bowel Dis 2018;24(7):1547–1557. DOI: 10.1093/ibd/izy046.
135. Feng YH, Tsao CJ. Emerging role of microRNA-21 in cancer. Biomed Rep 2016;5(4):395–402. DOI: 10.3892/br.2016.747.
136. Hou J, Hu X, Chen B, et al. miR-155 targets Est-1 and induces ulcerative colitis via the IL-23/17/6-mediated Th17 pathway. Pathol Res Practice 2017;213(10):1289–1295. DOI: 10.1016/j.prp.2017.08.001.
137. Paraskevi A, Theodoropoulos G, Papaconstantinou I, et al. Circulating MicroRNA in inflammatory bowel disease. J Crohns Colitis 2012;6(9):900–904. DOI: 10.1016/j.crohns.2012.02.006.
138. Shibuya H, Iinuma H, Shimada R, et al. Clinicopathological and prognostic value of microRNA-21 and microRNA-155 in colorectal cancer. Oncology 2010;79(3–4):313–320. DOI: 10.1159/000323283.
139. Qiao Y, Cai CW, Shen J, et al. Circular RNA expression alterations in colon tissues of Crohn’s disease patients. Mol Med Rep 2019;19(5):4500–4506. DOI: 10.3892/mmr.2019.10070.
140. Wang T, Chen N, Ren W, et al. Integrated analysis of circRNAs and mRNAs expression profile revealed the involvement of hsa_circ_0007919 in the pathogenesis of ulcerative colitis. J Gastroenterol 2019;54(9):804–818. DOI: 10.1007/s00535-019-01585-7.
141. Yin J, Hu T, Xu L, et al. Circular RNA expression profile in peripheral blood mononuclear cells from Crohn disease patients. Medicine (Baltimore) 2019;98(26):e16072. DOI: 10.1097/MD.0000000000016072.
142. Xie Y, Zhao Y, Shi L, et al. Gut epithelial TSC1/mTOR controls RIPK3-dependent necroptosis in intestinal inflammation and cancer. J Clin Invest 2020;130(4):2111–2128. DOI: 10.1172/JCI133264.
143. Slowicka K, Serramito-Gómez I, Boada-Romero E, et al. Physical and functional interaction between A20 and ATG16L1-WD40 domain in the control of intestinal homeostasis. Nat Commun 2019;10(1):1834. DOI: 10.1038/s41467-019-09667-z.
144. Cuna A, George L, Sampath V. Genetic predisposition to necrotizing enterocolitis in premature infants: current knowledge, challenges, and future directions. Semin Fetal Neonatal Med 2018;23(6):387–393. DOI: 10.1016/j.siny.2018.08.006.
145. Sampath V, Bhandari V, Berger J, et al. A functional ATG16L1 (T300A) variant is associated with necrotizing enterocolitis in premature infants. Pediatr Res 2017;81(4):582–588. DOI: 10.1038/pr.2016.260.
146. Li XX, Xiao L, Chung HK, et al. Interaction between HuR and circPABPN1 modulates autophagy in the intestinal epithelium by altering ATG16L1 translation. Mol Cell Biol 2020;40(6):e00492-19. DOI: 10.1128/MCB.00492-19.
147. Ye YL, Yin J, Hu T, et al. Increased circulating circular RNA_103516 is a novel biomarker for inflammatory bowel disease in adult patients. World J Gastroenterol 2019;25(41):6273–6288. DOI: 10.3748/wjg.v25.i41.6273.
148. Lin L, Zhou G, Chen P, et al. Which long noncoding RNAs and circular RNAs contribute to inflammatory bowel disease? Cell Death Dis 2020;11(6):456. DOI: 10.1038/s41419-020-2657-z.
149. Zou Y, Jiang Z, Yu X, et al. Upregulation of long noncoding RNA SPRY4-IT1 modulates proliferation, migration, apoptosis, and network formation in trophoblast cells HTR-8SV/neo. PLoS One 2013;8(11):e79598. DOI: 10.1371/journal.pone.0079598.
150. Liu R, Tang A, Wang X, et al. Inhibition of lncRNA NEAT1 suppresses the inflammatory response in IBD by modulating the intestinal epithelial barrier and by exosome-mediated polarization of macrophages. Int J Mol Med 2018;42(5):2903–2913. DOI: 10.3892/ijmm.2018.3829.
151. Xiao L, Rao JN, Cao S, et al. Long noncoding RNA SPRY4-IT1 regulates intestinal epithelial barrier function by modulating the expression levels of tight junction proteins. Mol Biol Cell 2016;27(4):617–626. DOI: 10.1091/mbc.E15-10-0703.
152. Mirza AH, Berthelsen CHB, Seemann SE, et al. Transcriptomic landscape of lncRNAs in inflammatory bowel disease. Genome Med 2015;7(1):39. DOI: 10.1186/s13073-015-0162-2.
153. Qiao Y, Huang ML, Xu AT, et al. LncRNA DQ786243 affects Treg related CREB and Foxp3 expression in Crohn’s disease. J Biomed Sci 2013;20(1):87. DOI: 10.1186/1423-0127-20-87.
154. Quan Y, Song K , Zhang Y, et al. Roseburia intestinalis -derived flagellin is a negative regulator of intestinal inflammation. Biochem Biophys Res Commun 2018;501(3):791–799. DOI: 10.1016/j.bbrc.2018.05.075.
155. Qiao C, Yang L, Wan J, et al. Long noncoding RNA ANRIL contributes to the development of ulcerative colitis by miR-323b-5p/TLR4/MyD88/NF-кB pathway. Biochem Biophys Res Commun 2019;508(1):217–224. DOI: 10.1016/j.bbrc.2018.11.100.
________________________
© The Author(s). 2022 Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (https://creativecommons.org/licenses/by-nc/4.0/), which permits unrestricted use, distribution, and non-commercial reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.