Circular RNA expression alterations in colon tissues of Crohn's disease patients

  • Authors:
    • Yu Qi Qiao
    • Chen Wen Cai
    • Jun Shen
    • Qing Zheng
    • Zhi Hua Ran
  • View Affiliations

  • Published online on: March 21, 2019     https://doi.org/10.3892/mmr.2019.10070
  • Pages: 4500-4506
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Genetic factors are crucial in the development of Crohn's disease (CD). Circular RNAs (circRNAs) are known to function as microRNA (miRNA) sponges and regulate a number of signalling pathways via circRNA‑miRNA interactions. As competing endogenous RNAs, the functions of circRNAs in CD should be investigated. In the present study, colon biopsy tissues were collected from ileocolon (L3)‑active CD patients and healthy controls. circRNA microarrays were performed with colon tissues from 3 CD patients and 3 controls. Subsequently, the candidate circRNAs were verified via reverse transcription‑quantitative polymerase chain reaction using colon tissues from a further 10 CD patients and 10 controls. Targeted miRNAs, genes and pathways of candidate circRNAs were predicted and analysed. Arraystar circRNA microarrays demonstrated that there were 163 upregulated circRNAs targeting 435 miRNAs and 55 downregulated circRNAs targeting 207 miRNAs (fold‑change >2 and P<0.01) in CD patients. As a candidate circRNA, hsa‑circRNA‑102685 was observed to putatively target hsa‑miR‑146b‑5p, hsa‑miR‑182‑5p and hsa‑miR‑146a‑5p. Furthermore, Kyoto Encyclopaedia of Genes and Genomes pathway analysis predicted that hsa‑circRNA‑102685 potentially participated in apoptosis, and in the Toll‑like receptor and p53 signalling pathways. Overall, the current study suggested that circRNA alterations serve an important role in the pathogenesis of CD. circRNAs, such as hsa‑circRNA‑102685, are involved in certain important signalling pathways of CD, and may be novel targets for diagnosis or treatment in this disease.

Introduction

Inflammatory bowel disease (IBD) refers to a group of conditions characterised by chronic inflammation of the bowel, including Crohn's disease (CD) and ulcerative colitis. A number of factors are associated with the development of IBD, including genetic, environmental, microbial and immune response factors (1,2). It has been reported that microRNAs (miRNAs) serve important roles in IBD (37). For instance, several studies have demonstrated an association between miR-21 and IBD in patients and animal models (8,9). In addition, Brain et al (10) provided a profile of how nucleotide-binding oligomerization domain-containing protein 2 (NOD2) induced miR-29 to limit interleukin-23 release in human dendritic cells (DCs). NOD2 mutations have also been reported to be associated with CD (11). Brest et al (12) identified that a variant in immunity-related GTPase family M protein (IRGM) gene altered a binding site for miR-196 and caused the deregulation of IRGM-dependent xenophagy in CD. In a murine model of IBD, miR-146 regulated NOD2-derived gut inflammation and served an important role in amplifying inflammatory responses (13). Other functions of miR-146 were also confirmed to be associated with IBD, including the regulation of the Toll-like receptor (TLR) signalling pathways, T cell differentiation and regulator T (Treg) cell function (7,14).

Circular RNA (circRNA) is a type of RNA that can form a covalently closed loop. The 5′ cap or 3′ poly(A) tail are not found in circRNA, and this characteristic results in the difference of circRNAs from other linear RNAs. In the 1970s, circRNAs were detected in viruses and extracted from certain eukaryotic cells; however, their functions were not fully understood at that time (15,16). In 2010, Poliseno et al (17) demonstrated that expressed pseudogenes were able to regulate the expression of coding genes. Salmena et al (18) further hypothesised that circRNAs are competing endogenous RNAs (ceRNAs). This hypothesis suggested that miRNAs do not interact with messenger RNAs (mRNAs) unidirectionally, but they interact with the pool of mRNAs, transcribed pseudogenes and long noncoding RNAs (18). In 2013, Memczak et al (19) described circRNAs as a large class of RNAs with regulatory potency, with thousands of circRNAs potentially existing stably in eukaryotic cells. These authors further revealed that the human circRNA CDR1as was able to be densely bound by miRNA (19). circRNAs reportedly function as miRNA sponges, while they have not been found to function as coding proteins to date. Currently, the most likely role of circRNAs is considered to be as ceRNAs.

Although miRNAs are known to serve important roles in the pathogenesis of IBD, the roles of circRNAs in the development of IBD remain unclear. The present study investigated the expression levels of circRNAs, considered as miRNA sponges, in the colonic tissues of CD patients. In addition, one of the potential circRNA-miRNA-gene networks was predicted by further examining a circRNA candidate.

Materials and methods

Patients and samples

Between April 2015 and March 2017, 13 patients with active CD (8 males and 5 females, mean age 30.69±6.25, from 18 to 38, without any other known diseases) who were treated at the Renji Hospital (Shanghai, China) and 13 healthy controls (7 males and 6 females, mean age 32.77±5.89, from 24 to 43, without any other known diseases), who were physicians at this hospital, were enrolled into the present study. The diagnosis of CD was established based on the medical history, endoscopy, biopsy, imaging and serological studies. The diagnosis of CD was confirmed by two different gastroenterologists. All patients were classified into the L3 (ileocolon) type of CD according to the Montreal Classification, and exhibited a Harvey-Bradshaw index score of ≥5 (20,21). All colonic samples were collected under colonoscopy prior to treatment. Samples were stored in RNAlater stabilization reagent (Qiagen, Valencia, CA, USA) following the protocol provided by the manufacturer. Samples from 3 random patients with active CD and 3 random healthy controls were initially selected for circRNA microarray analysis. The remaining samples from 10 CD patients and 10 healthy controls were used for reverse transcription-quantitative polymerase chain reaction (RT-qPCR) verification. All patients and healthy donors were well informed of the study details, and provided written informed consent. The experiments were approved by the Ethics Committee of Renji Hospital, School of Medicine, Shanghai Jiao Tong University (Shanghai, China).

RNA isolation

Total RNA was extracted from the colonic samples using TRIzol™ reagent (Invitrogen; Thermo Fisher Scientific, Inc., Waltham, MA, USA) following the protocol provided by the manufacturer. Next, RNA was purified with the RNeasy Mini kit (Qiagen) according to the manufacturer's guidelines. A NanoDrop ND-1000 spectrophotometer (Thermo Fisher Scientific, Inc.) was used to measure the concentration of RNA based on the OD260/OD280 ratio.

circRNA microarray analysis

Arraystar Human Circular RNA Array (Arraystar, Inc., Rockville, MD, USA) analysis was respectively performed in 3 samples from patients and controls to determine the expression profile of circRNAs. The sample preparation and microarray hybridization were performed based on the standard protocols provided by Arraystar. Briefly, total RNA from each sample was amplified and transcribed into fluorescent complementary RNA utilizing random primers, according to the Arraystar Super RNA Labelling protocol (Arraystar, Inc.). The labelled circRNAs were hybridised onto the Arraystar Human circRNA Array (6×7K; Arraystar, Inc.). Subsequent to washing the slides, the arrays were scanned by the Axon GenePix 4000B microarray scanner (Molecular Devices, LLC, Sunnyvale, CA, USA). The scanned images were then imported into the Axon GenePix Pro 6.0 software (Molecular Devices, LLC) for grid alignment and data extraction. The R package (https://www.r-project.org/) was used for quantile normalization and subsequent expression matrix processing. Differentially expressed circRNAs between the two groups that presented a statistically significant variance were confirmed by filtering with a volcano plot. For the selection of circRNA candidates, differentially expressed circRNAs were defined as having a fold-change of >2 and P<0.01. Hierarchical clustering was then performed to identify distinguishable patterns of circRNA expression among the samples. Since miR-146 has already been demonstrated to be involved in the pathogenesis of CD (7), one of the circRNAs that putatively targeted both miR-146a and miR-146b was selected as the candidate.

RT-qPCR

To validate the circRNA expression profile detected using the Arraystar Human circRNA Array, the expression level of hsa-circRNA-102685 (circRNA-102685) was further validated by RT-qPCR in the remaining samples from 10 CD patients and 10 healthy controls. Briefly, total RNA was incubated for 1 h at 37°C with 0 units (serving as the mock treatment) or 20 units of RNase R (Epicentre; Illumina, Inc., San Diego, CA, USA) (22). RNA was subsequently purified with the RNeasy Mini kit (Qiagen), and reverse transcribed to synthesise cDNA with ReverTra Ace qPCR RT Kit (Toyobo, Osaka, Japan). The process of reverse transcription was performed according with the manufacturer's instructions: Incubation of RNA solution at 65°C for 5 min, followed by incubation at 37°C for 15 min, and lastly heating at 98°C for 5 min. Next, the AFD9600 Real-time PCR Detection system (Hangzhou AGS MedTech Co., Ltd., Hangzhou, China) and the SYBR Green qPCR SuperMix (Invitrogen; Thermo Fisher Scientific, Inc.) were applied to perform qPCR assay. Primers were designed using Primer Premier 6.0 (Premier Biosoft International, Palo Alto, CA, USA), and were as follows: circRNA-102685, 5′-ACATTCTGCCAGTGCGTCAAC-3′ (forward) and 5′-GAATCTTCAGGACAACGTGGAGAG-3′ (reverse); GAPDH, 5′-ACTTTGGTATCGTGGAAGGACTCAT-3′ (forward) and 5′-GTTTTTCTAGACGGCAGGTCAGG-3′ (reverse). GAPDH served as an internal standard control. The PCR conditions included a 95°C denaturation for 5 min, followed by 40 cycles at 95°C for 5 sec and 61°C for 30 sec. Three independent samples were tested for each CD patient and control individual, and all reactions were performed in triplicate. The quantification cycle (Cq) was applied to define the expression of circRNA-102685, and the 2−ΔΔCq method was used to calculate the relative expression levels (23).

Bioinformatics analysis and biological function prediction

Arraystar's homemade miRNA target prediction software based on TargetScan (24) and miRanda (25) was applied for the prediction of circRNA-miRNA interactions. The top 5 potential target miRNAs were selected from the analysis. IBD-associated miRNAs were selected based on the review published by Kalla et al (7), while TargetScan (http://www.targetscan.org) was used to predict the putative target genes of miRNAs. A visualised circRNA-miRNA-gene network was subsequently generated using the Cytoscape software platform (https://cytoscape.org/). Gene Ontology (GO) enrichment analysis was conducted for annotating the functions of the target genes, while Kyoto Encyclopaedia of Genes and Genomes (KEGG) pathway enrichment analysis was also performed to determine the involved pathways (2628).

Statistical analysis

Student's t-test was used for continuous variables, and data are expressed as the mean ± standard deviation. P<0.05 was considered to indicate a statistically significant difference. Statistical analyses were performed using R package (version 3.4.4) and GraphPad Prism software (version 5.0; GraphPad Software, Inc., La Jolla, CA, USA).

Results

circRNA expression profiling in colon tissues of CD

Overall, 218 circRNAs were identified to be differentially expressed in colon tissues between CD patients and controls. A total of 163 circRNAs were upregulated in the CD group, and these putatively targeted 435 miRNAs. In addition, 55 circRNAs were downregulated in the CD group and putatively targeted 207 miRNAs. Hierarchical clustering and patterns of circRNA expression are displayed in Fig. 1. Of these putative miR-146-targeting circRNAs, 4 were upregulated and 3 were downregulated (Table I). circRNA-102685 was the only upregulated circRNA that putatively targeted both miR-146a and miR-146b, and was thus selected for further investigation.

Table I.

Differentially expressed circRNAs that putatively target microRNA-146 in Crohn's disease.

Table I.

Differentially expressed circRNAs that putatively target microRNA-146 in Crohn's disease.

circRNAFold changeP-value
Upregulated
  hsa_circRNA_1027748.564309<0.001
  hsa_circRNA_1017537.1439330.006
  hsa_circRNA_1026853.0206090.004
  hsa_circRNA_1022614.532291<0.001
Downregulated
  hsa_circRNA_1015983.315920.008
  hsa_circRNA_1018632.4241990.003
  hsa_circRNA_1001892.3370420.002

[i] circRNA, circular RNA.

circRNA candidate selection and validation with RT-qPCR

According to the aforementioned results, circRNA-102685 was selected as a candidate for further verification. In the circRNA microarray analysis, the genomic locus of circRNA-102685 was detected to be on chromosome 2, and the predicted best transcript was uc002rpd.3. Furthermore, circRNA-102685 was upregulated by 3.02-fold in the CD group as compared with the control group (P<0.01). According to the results of RT-qPCR validation, circRNA-102685 was upregulated by 1.46-fold in the CD group as compared with the control group (P<0.01; Fig. 2).

Candidate circRNA-miRNA-gene network prediction

As circRNAs are potential upstream molecular sponges for their target miRNAs, the TargetScan and miRanda tools were used to predict the circRNA-miRNA-gene network. The top 3 putative miRNA targets of circRNA-102685 were hsa-miR-146b-5p, hsa-miR-182-5p and hsa-miR-146a-5p. In total, 47 target genes of these 3 miRNAs were identified by target gene prediction using TargetScan database. Next, the circRNA-102685-miRNA-gene network was constructed, and is displayed in Fig. 3. Subsequent prediction of biological pathways provided a profile of the potential functions of these target genes. The target genes were strongly associated with KEGG pathways that were associated with cancer (including pathways in cancer, prostate cancer, pancreatic cancer, melanoma, chronic myeloid leukaemia, colorectal cancer and small cell lung cancer), cell growth and death (including apoptosis, cell cycle, and p53 signalling pathways), immune system (including TLR, RIG-I-like receptor and chemokine signalling pathways), nervous system (namely neurotrophin signalling pathway), cellular community-eukaryotes (adherens junction), endocrine system (melanogenesis), transport and catabolism (endocytosis), and signalling molecules and interaction (cytokine-cytokine receptor interaction). The significant KEGG pathways are displayed in Fig. 4A. Furthermore, GO analysis revealed that these target genes were mainly associated with cell death, apoptosis and phosphorylation (Fig. 4B).

Discussion

CD is a life-long disease involving chronic gastrointestinal inflammation; however, its causes are currently not fully understood. As the functions of miRNAs have been reported to be associated with CD (7), the current study aimed to examine the profile of circRNAs in this disease, which are another type of RNAs. circRNAs can function as miRNA sponges, and may work together with miRNAs to regulate the expression of target genes (29). In addition, circRNAs are very stable in eukaryotic cells, with a half-life that can reach 48 h, while the half-life of miRNAs is only 10 h (30). This characteristic makes circRNAs stable molecular markers in diseases.

circRNAs have been reported to be potential molecular markers in different types of cancer (31). However, a number of studies have demonstrated that circRNAs may also be involved in certain non-cancer diseases. For instance, Wang et al (32) observed that a heart-associated circRNA targeting miR-223 was able to protect the heart, and serve as a potential therapeutic target in the treatment of cardiac hypertrophy and heart failure. Furthermore, the circRNA CDR1as was reported to be associated with neurodegenerative conditions (33). In chronic autoimmune disease, circRNAs are differentially expressed. The study by Liu et al (34) indicated that circ-CER may be associated with degenerative changes in the joint cartilage, which can be a potential target in osteoarthritis.

As a type of chronic autoimmune disease, CD was investigated in the present study, and alterations in the expression of circRNAs in CD patients were examined. The results demonstrated that >200 circRNAs were differentially expressed between CD patients and healthy controls. These circRNAs may function as sponges of miRNAs and serve different roles in the pathogenesis of CD.

Among the differentially expressed circRNAs identified in the current study, 7 candidates putatively targeted miR-146. Previously, miR-146 has been reported to be involved in the TLR signalling pathway, and thus serves a role in the pathogenesis of CD (35). Taganov et al (35) identified that the expression of both miR-146a and miR-146b in monocytes was induced by lipopolysaccharide, flagellin, peptidoglycan and exposure to TLR ligands. In animal experiments, Nata et al (36) demonstrated that miR-146b improved intestinal inflammation by activating nuclear factor-κB. Furthermore, in a small sample study, Fasseu et al (37) identified that miR-146 was upregulated in CD tissues compared with the controls. A review by Xu and Zhang (5) summarised that miR-146 may affect the differentiation, maturation and functions of several different immune cells, including DCs, helper T cells and Treg cells. The present study identified that circRNA-102685 was upregulated by performing a circRNA array analysis, and that it potentially targeted both miR-146a and miR-146b. Thus, circRNA-102685 was selected as a candidate circRNA for further validation by RT-qPCR. The RT-qPCR results demonstrated that circRNA-102685 was upregulated in colon tissues obtained from CD patients as compared with the healthy controls. Therefore, circRNA-102685 may serve a role in regulating target gene expression via miR-146 in the pathogenesis of CD.

Further investigation focused on the potential circRNA-miRNA-gene network. In the current study, it was predicted that circRNA-102685 potentially functioned as a sponge of miR-146. In the predicted circRNA-miRNA-gene network, a number of important genes were identified that are known to be associated with the pathogenesis of CD, such as IRAK1, IRAK2, BCL2, NFκB1, CREB1 and TRAF6 (3842). Furthermore, the prediction of KEGG pathways presented several top pathways associated with circRNA-102685, including apoptosis, TLR and p53 signalling pathways. These pathways have been demonstrated to be associated with IBD (4346). Other pathways, such as the RIG-I-like receptor signalling pathway, chemokine signalling pathway and cytokine-cytokine receptor interaction pathway, are potentially associated with IBD (4749). The association between cancer and IBD has already been demonstrated (50), and the prediction conducted in the current study identified the involvement of certain cancer-associated pathways. GO enrichment analysis presented similar results. However, the role of circRNA-102685 in these pathways requires further research.

There are several limitations in the current study. As the size and number of biopsy tissues were limited in patients with active CD, it was not possible to verify the expression of other circRNAs. In addition, only 13 CD patients and 13 controls were enrolled in the study. More studies on the role of circRNAs in IBD should be performed in the future.

In conclusion, the present study revealed that circRNAs were differentially expressed in colon tissues of CD patients in comparison with the healthy controls. circRNA expression alterations may serve an important role in the pathogenesis of CD. The expression changes of hsa-circRNA-102685 may be involved in the process of CD. Certain associated pathways that may be involved in the process of CD were identified, and these may serve as novel targets in the diagnosis and treatment of this disease.

Acknowledgements

The authors would like to thank Dr Si Bo Zhu (Fudan University) for his help in the bioinformatics analysis.

Funding

This research was supported by the National Natural Science Foundation of China (grant nos. 81500424 and 81670497).

Availability of data and materials

The datasets used and/or analysed during the current study are available from the corresponding author upon reasonable request.

Authors' contributions

YQQ and JS designed the study. YQQ worked on the bioinformatics analysis and drafted the manuscript. CWC collected the samples and performed the verification work. QZ and ZHR co-worked on the analysis and critically reviewed the interpretation of data. All authors read and approved the final version of the manuscript.

Ethics approval and consent to participate

All patients and healthy donors were well informed of the study details, and provided written informed consent. The experiments were approved by the Ethics Committee of Renji Hospital, School of Medicine, Shanghai Jiao Tong University.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Lee SH, Kwon JE and Cho ML: Immunological pathogenesis of inflammatory bowel disease. Intest Res. 16:26–42. 2018. View Article : Google Scholar : PubMed/NCBI

2 

de Mattos BR, Garcia MP, Nogueira JB, Paiatto LN, Albuquerque CG, Souza CL, Fernandes LG, Tamashiro WM and Simioni PU: Inflammatory bowel disease: An overview of immune mechanisms and biological treatments. Mediators Inflamm. 2015:4930122015. View Article : Google Scholar : PubMed/NCBI

3 

Dalal SR and Kwon JH: The role of MicroRNA in inflammatory bowel disease. Gastroenterol Hepatol (NY). 6:714–722. 2010.

4 

Chapman CG and Pekow J: The emerging role of miRNAs in inflammatory bowel disease: A review. Therap Adv Gastroenterol. 8:4–22. 2015. View Article : Google Scholar : PubMed/NCBI

5 

Xu XM and Zhang HJ: miRNAs as new molecular insights into inflammatory bowel disease: Crucial regulators in autoimmunity and inflammation. World J Gastroenterol. 22:2206–2218. 2016. View Article : Google Scholar : PubMed/NCBI

6 

Schaefer JS, Attumi T, Opekun AR, Abraham B, Hou J, Shelby H, Graham DY, Streckfus C and Klein JR: MicroRNA signatures differentiate Crohn's disease from ulcerative colitis. BMC Immunol. 16:52015. View Article : Google Scholar : PubMed/NCBI

7 

Kalla R, Ventham NT, Kennedy NA, Quintana JF, Nimmo ER, Buck AH and Satsangi J: MicroRNAs: New players in IBD. Gut. 64:504–517. 2015. View Article : Google Scholar : PubMed/NCBI

8 

Adams AT, Kennedy NA, Hansen R, Ventham NT, O'Leary KR, Drummond HE, Noble CL, El-Omar E, Russell RK, Wilson DC, et al: Two-stage genome-wide methylation profiling in childhood-onset Crohn's disease implicates epigenetic alterations at the VMP1/MIR21 and HLA loci. Inflamm Bowel Dis. 20:1784–1793. 2014. View Article : Google Scholar : PubMed/NCBI

9 

Wu F, Dong F, Arendovich N, Zhang J, Huang Y and Kwon JH: Divergent influence of microRNA-21 deletion on murine colitis phenotypes. Inflamm Bowel Dis. 20:1972–1985. 2014. View Article : Google Scholar : PubMed/NCBI

10 

Brain O, Owens BM, Pichulik T, Allan P, Khatamzas E, Leslie A, Steevels T, Sharma S, Mayer A, Catuneanu AM, et al: The intracellular sensor NOD2 induces microRNA-29 expression in human dendritic cells to limit IL-23 release. Immunity. 39:521–536. 2013. View Article : Google Scholar : PubMed/NCBI

11 

Wehkamp J, Harder J, Weichenthal M, Schwab M, Schäffeler E, Schlee M, Herrlinger KR, Stallmach A, Noack F, Fritz P, et al: NOD2 (CARD15) mutations in Crohn's disease are associated with diminished mucosal alpha-defensin expression. Gut. 53:1658–1664. 2004. View Article : Google Scholar : PubMed/NCBI

12 

Brest P, Lapaquette P, Souidi M, Lebrigand K, Cesaro A, Vouret-Craviari V, Mari B, Barbry P, Mosnier JF, Hébuterne X, et al: A synonymous variant in IRGM alters a binding site for miR-196 and causes deregulation of IRGM-dependent xenophagy in Crohn's disease. Nat Genet. 43:242–245. 2011. View Article : Google Scholar : PubMed/NCBI

13 

Ghorpade DS, Sinha AY, Holla S, Singh V and Balaji KN: NOD2-nitric oxide-responsive microRNA-146a activates Sonic hedgehog signaling to orchestrate inflammatory responses in murine model of inflammatory bowel disease. J Biol Chem. 288:33037–33048. 2013. View Article : Google Scholar : PubMed/NCBI

14 

He X, Jing Z and Cheng G: MicroRNAs: New regulators of Toll-like receptor signalling pathways. Biomed Res Int. 2014:9451692014. View Article : Google Scholar : PubMed/NCBI

15 

Sanger HL, Klotz G, Riesner D, Gross HJ and Kleinschmidt AK: Viroids are single-stranded covalently closed circular RNA molecules existing as highly base-paired rod-like structures. Proc Natl Acad Sci USA. 73:3852–3856. 1976. View Article : Google Scholar : PubMed/NCBI

16 

Hsu MT and Coca-Prados M: Electron microscopic evidence for the circular form of RNA in the cytoplasm of eukaryotic cells. Nature. 280:339–340. 1979. View Article : Google Scholar : PubMed/NCBI

17 

Poliseno L, Salmena L, Zhang J, Carver B, Haveman WJ and Pandolfi PP: A coding-independent function of gene and pseudogene mRNAs regulates tumour biology. Nature. 465:1033–1038. 2010. View Article : Google Scholar : PubMed/NCBI

18 

Salmena L, Poliseno L, Tay Y, Kats L and Pandolfi PP: A ceRNA hypothesis: The Rosetta stone of a hidden RNA language? Cell. 146:353–358. 2011. View Article : Google Scholar : PubMed/NCBI

19 

Memczak S, Jens M, Elefsinioti A, Torti F, Krueger J, Rybak A, Maier L, Mackowiak SD, Gregersen LH, Munschauer M, et al: Circular RNAs are a large class of animal RNAs with regulatory potency. Nature. 495:333–338. 2013. View Article : Google Scholar : PubMed/NCBI

20 

Harvey RF and Bradshaw JM: A simple index of Crohn's-disease activity. Lancet. 1:5141980. View Article : Google Scholar : PubMed/NCBI

21 

Satsangi J, Silverberg MS, Vermeire S and Colombel JF: The Montreal classification of inflammatory bowel disease: Controversies, consensus, and implications. Gut. 55:749–753. 2006. View Article : Google Scholar : PubMed/NCBI

22 

Li L, Guo J, Chen Y, Chang C and Xu C: Comprehensive CircRNA expression profile and selection of key CircRNAs during priming phase of rat liver regeneration. BMC Genomics. 18:802017. View Article : Google Scholar : PubMed/NCBI

23 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

24 

Enright AJ, John B, Gaul U, Tuschl T, Sander C and Marks DS: MicroRNA targets in Drosophila. Genome Biol. 5:R12003. View Article : Google Scholar : PubMed/NCBI

25 

Pasquinelli AE: MicroRNAs and their targets: Recognition, regulation and an emerging reciprocal relationship. Nat Rev Genet. 13:271–282. 2012. View Article : Google Scholar : PubMed/NCBI

26 

Kanehisa M, Furumichi M, Tanabe M, Sato Y and Morishima K: KEGG: New perspectives on genomes, pathways, diseases and drugs. Nucleic Acids Res. 45:D353–D361. 2017. View Article : Google Scholar : PubMed/NCBI

27 

Ashburner M, Ball CA, Blake JA, Botstein D, Butler H, Cherry JM, Davis AP, Dolinski K, Dwight SS, Eppig JT, et al: Gene ontology: Tool for the unification of biology. The Gene Ontology Consortium. Nat Genet. 25:25–29. 2000. View Article : Google Scholar : PubMed/NCBI

28 

The Gene Ontology Consortium: Expansion of the gene ontology knowledgebase and resources. Nucleic Acids Res. 45:D331–D338. 2017. View Article : Google Scholar : PubMed/NCBI

29 

Hansen TB, Jensen TI, Clausen BH, Bramsen JB, Finsen B, Damgaard CK and Kjems J: Natural RNA circles function as efficient microRNA sponges. Nature. 495:384–388. 2013. View Article : Google Scholar : PubMed/NCBI

30 

Jeck WR and Sharpless NE: Detecting and characterizing circular RNAs. Nat Biotechnol. 32:453–461. 2014. View Article : Google Scholar : PubMed/NCBI

31 

Meng S, Zhou H, Feng Z, Xu Z, Tang Y, Li P and Wu M: CircRNA: Functions and properties of a novel potential biomarker for cancer. Mol Cancer. 16:942017. View Article : Google Scholar : PubMed/NCBI

32 

Wang K, Long B, Liu F, Wang JX, Liu CY, Zhao B, Zhou LY, Sun T, Wang M, Yu T, et al: A circular RNA protects the heart from pathological hypertrophy and heart failure by targeting miR-223. Eur Heart J. 37:2602–2611. 2016. View Article : Google Scholar : PubMed/NCBI

33 

Lukiw WJ: Circular RNA (circRNA) in Alzheimer's disease (AD). Front Genet. 4:3072013. View Article : Google Scholar : PubMed/NCBI

34 

Liu Q, Zhang X, Hu X, Dai L, Fu X, Zhang J and Ao Y: Circular RNA related to the chondrocyte ECM regulates MMP13 expression by functioning as a MiR-136 ‘Sponge’ in human cartilage degradation. Sci Rep. 6:225722016. View Article : Google Scholar : PubMed/NCBI

35 

Taganov KD, Boldin MP, Chang KJ and Baltimore D: NF-kappaB-dependent induction of microRNA miR-146, an inhibitor targeted to signaling proteins of innate immune responses. Proc Natl Acad Sci USA. 103:12481–12486. 2006. View Article : Google Scholar : PubMed/NCBI

36 

Nata T, Fujiya M, Ueno N, Moriichi K, Konishi H, Tanabe H, Ohtake T, Ikuta K and Kohgo Y: MicroRNA-146b improves intestinal injury in mouse colitis by activating nuclear factor-kappaB and improving epithelial barrier function. J Gene Med. 15:249–260. 2013. View Article : Google Scholar : PubMed/NCBI

37 

Fasseu M, Tréton X, Guichard C, Pedruzzi E, Cazals-Hatem D, Richard C, Aparicio T, Daniel F, Soulé JC, Moreau R, et al: Identification of restricted subsets of mature microRNA abnormally expressed in inactive colonic mucosa of patients with inflammatory bowel disease. PLoS One. 5:e131602010. View Article : Google Scholar : PubMed/NCBI

38 

Zhernakova A, Festen EM, Franke L, Trynka G, van Diemen CC, Monsuur AJ, Bevova M, Nijmeijer RM, van 't Slot R, Heijmans R, et al: Genetic analysis of innate immunity in Crohn's disease and ulcerative colitis identifies two susceptibility loci harboring CARD9 and IL18RAP. Am J Hum Genet. 82:1202–1210. 2008. View Article : Google Scholar : PubMed/NCBI

39 

Catarzi S, Marcucci T, Papucci L, Favilli F, Donnini M, Tonelli F, Vincenzini MT and Iantomasi T: Apoptosis and Bax, Bcl-2, Mcl-1 expression in neutrophils of Crohn's disease patients. Inflamm Bowel Dis. 14:819–825. 2008. View Article : Google Scholar : PubMed/NCBI

40 

Sartor RB: Mechanisms of disease: Pathogenesis of Crohn's disease and ulcerative colitis. Nat Clin Pract Gastroenterol Hepatol. 3:390–407. 2006. View Article : Google Scholar : PubMed/NCBI

41 

Diegelmann J, Czamara D, Le Bras E, Zimmermann E, Olszak T, Bedynek A, Göke B, Franke A, Glas J and Brand S: Intestinal DMBT1 expression is modulated by Crohn's disease-associated IL23R variants and by a DMBT1 variant which influences binding of the transcription factors CREB1 and ATF-2. PLoS One. 8:e777732013. View Article : Google Scholar : PubMed/NCBI

42 

Shen J, Qiao Y, Ran Z and Wang T: Different activation of TRAF4 and TRAF6 in inflammatory bowel disease. Mediators Inflamm. 2013:6479362013. View Article : Google Scholar : PubMed/NCBI

43 

Nunes T, Bernardazzi C and de Souza HS: Cell death and inflammatory bowel diseases: Apoptosis, necrosis, and autophagy in the intestinal epithelium. Biomed Res Int. 2014:2184932014. View Article : Google Scholar : PubMed/NCBI

44 

Mudter J and Neurath MF: Apoptosis of T cells and the control of inflammatory bowel disease: Therapeutic implications. Gut. 56:293–303. 2007. View Article : Google Scholar : PubMed/NCBI

45 

Cario E: Toll-like receptors in inflammatory bowel diseases: A decade later. Inflamm Bowel Dis. 16:1583–1597. 2010. View Article : Google Scholar : PubMed/NCBI

46 

Goretsky T, Dirisina R, Sinh P, Mittal N, Managlia E, Williams DB, Posca D, Ryu H, Katzman RB and Barrett TA: p53 mediates TNF-induced epithelial cell apoptosis in IBD. Am J Pathol. 181:1306–1315. 2012. View Article : Google Scholar : PubMed/NCBI

47 

Ireton RC and Gale M Jr: RIG-I like receptors in antiviral immunity and therapeutic applications. Viruses. 3:906–919. 2011. View Article : Google Scholar : PubMed/NCBI

48 

Singh UP, Singh NP, Murphy EA, Price RL, Fayad R, Nagarkatti M and Nagarkatti PS: Chemokine and cytokine levels in inflammatory bowel disease patients. Cytokine. 77:44–49. 2016. View Article : Google Scholar : PubMed/NCBI

49 

Xavier RJ and Podolsky DK: Unravelling the pathogenesis of inflammatory bowel disease. Nature. 448:427–434. 2007. View Article : Google Scholar : PubMed/NCBI

50 

Terzić J, Grivennikov S, Karin E and Karin M: Inflammation and colon cancer. Gastroenterology. 138:2101–2114.e5. 2010. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

May-2019
Volume 19 Issue 5

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Qiao YQ, Cai CW, Shen J, Zheng Q and Ran ZH: Circular RNA expression alterations in colon tissues of Crohn's disease patients. Mol Med Rep 19: 4500-4506, 2019
APA
Qiao, Y.Q., Cai, C.W., Shen, J., Zheng, Q., & Ran, Z.H. (2019). Circular RNA expression alterations in colon tissues of Crohn's disease patients. Molecular Medicine Reports, 19, 4500-4506. https://doi.org/10.3892/mmr.2019.10070
MLA
Qiao, Y. Q., Cai, C. W., Shen, J., Zheng, Q., Ran, Z. H."Circular RNA expression alterations in colon tissues of Crohn's disease patients". Molecular Medicine Reports 19.5 (2019): 4500-4506.
Chicago
Qiao, Y. Q., Cai, C. W., Shen, J., Zheng, Q., Ran, Z. H."Circular RNA expression alterations in colon tissues of Crohn's disease patients". Molecular Medicine Reports 19, no. 5 (2019): 4500-4506. https://doi.org/10.3892/mmr.2019.10070