Hindawi BioMed Research International Volume 2020, Article ID 7402657, 9 pages https://doi.org/10.1155/2020/7402657

Research Article Transcription Factors That Regulate the Pathogenesis of Ulcerative Colitis

Bing Zhang1 and Tao Sun 2

1TCM Department, Hangzhou First People’s Hospital, China 2Hepatology Department, The Second Affiliated Hospital of Zhejiang Chinese Medical University, 318th Chaowang Road, Gongshu District, Zhejiang, 310005 Hangzhou, China

Correspondence should be addressed to Tao Sun; [email protected]

Received 4 July 2020; Accepted 10 August 2020; Published 25 August 2020

Guest Editor: Tao Huang

Copyright © 2020 Bing Zhang and Tao Sun. This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Ulcerative colitis (UC) is one of the inflammatory bowel diseases (IBD) characterized by occurrence in the rectum and sigmoid colon of young adults. However, the functional roles of transcription factors (TFs) and their regulating target and pathways are not fully known in ulcerative colitis (UC). In this study, we collected expression data to identify differentially expressed TFs (DETFs). We found that differentially expressed genes (DEGs) were significantly enriched in the target genes of HOXA2, IKZF1, KLF2, XBP1, EGR2, ETV7, BACH2, CBFA2T3, HLF, and NFE2. TFs including BACH2, CBFA2T3, EGR2, ETV7, NFE2, and XBP1, and their target genes were significantly enriched in signaling by interleukins. BACH2 target genes were enriched in estrogen - (ESR-) mediated signaling and nongenomic estrogen signaling. Furthermore, to clarify the functional roles of immune cells on the UC pathogenesis, we estimated the immune cell proportions in all the samples. The accumulated effector CD8 and reduced proportion of naïve CD4 might be responsible for the adaptive immune response in UC. The accumulation of plasma in UC might be associated with increased gut permeability. In summary, we present a systematic study of the TFs by analyzing the DETFs, their regulating target genes and pathways, and immune cells. These findings might improve our understanding of the TFs in the pathogenesis of UC.

1. Introduction x`cancer by CNV arrays [4]. Moreover, FAM217B, KIAA1614, and RIBC2 were found to be hypermethylated Ulcerative colitis (UC) is one of the inflammatory bowel dis- in UC and could be used for the diagnosis and therapeutic eases (IBD) with symptoms such as abdominal pain, fever, treatment of UC based on genome-wide DNA methylation malnourishment, fatigue, and weight loss [1]. UC is charac- approach [5]. Furthermore, transcriptome-based system terized by occurrence in the rectum and sigmoid colon of biology approach identifies ANP32E, a involved in young adults aged 20-40 [2]. Currently, UC is recognized to steroid-refractoriness, indicating the key role of steroid- be caused by the damages of the intestinal mucosal barrier induced transcriptional changes and the implication of and neuroendocrine and immune dysfunction due to the ANP32E in UC [6]. In addition to these genes or , interplay of genetics, environment, and psychology [3], but miRNAs have been found to be implicated in UC. Particu- its specific etiology and pathogenesis are still unclear. larly, IL-33 expression was exerted by miR-378a-3p in an With the advances in high-throughput technologies, a inflammatory environment, and downregulation of miR- growing number of studies have been carried out to investi- 378a-3p could result in IL-33 overexpression in UC [7]. gate the expression of some genes and proteins in the patho- These studies greatly improved our understanding of the genesis and molecular mechanism of UC. Specifically, the underlying mechanism of UC pathogenesis. copy number variations (CNVs) in mitochondrial DNA have In addition, the transcription factors (TFs), a series of been identified as the predictor of UC-associated colorectal molecules involved in regulating , have been 2 BioMed Research International emerged as key regulators in several diseases [8, 9]. Heat 3. Results shock 2 could predict mucosal healing fi ff and promote mucosal repair by suppressing MAPK signaling 3.1. Identi cation of Di erentially Expressed Transcription and inhibit intestinal epithelial cell apoptosis in UC through Factors. The mucosal biopsies had 14 ulcerative colitis the mitochondrial pathway [10, 11]. Moreover, RUNX3 is (UC), 14 remission (R), and 16 healthy controls (N). With also associated with UC by regulating the immune-related the three groups of mucosal biopsies, we compared one with fi target genes and pathways [12, 13]. However, there is a lack the other two groups, respectively. UC had signi cantly dif- fi of systematic study analyzing the functional roles of TFs in ferent expression pro les as compared with R and N groups, ff the pathogenesis of UC. Therefore, we carried out the present with 3,202 and 2,517 di erentially expressed genes (DEGs) in study, aiming at identifying the critical TFs, their down- UC vs. N and UC vs. R (Supplementary Table S1), fi stream target genes, and pathways involved in UC respectively. The comparison of R vs. N only identi ed pathogenesis. 1,133 DEGs. Consistently, the comparisons of UC vs. N (n =56) and UC vs. R (n =46) had greater numbers of differentially expressed transcription factors (TFs) than that 2. Materials and Methods of R vs. N (n =6) (Figure 1(a)). These results indicated that the transcriptomic profiles were significantly altered in UC 2.1. Datasets. The gene expression data were collected from samples as compared with samples of remission and the Gene Expression Omnibus (GEO) database with acces- healthy controls. sion GSE128682, and the sample collection was described Totally, we identified 72 TFs significantly differentially in an earlier study [14]. The counts for each sample were nor- expressed between the three groups (Supplementary malized by DESeq2 [15]. The pairs of transcription factor Table S2). The hierarchical clustering analysis revealed that (TF) target genes were downloaded from three public data- the UC samples could be clearly differentiated from the N bases including JASPAR [16], TRANSFAC [17], and CHEA and R samples by the TFs specifically upregulated in UC [18]. (Figure 1(b)). The TFs specifically upregulated in R and N samples also had the capability of classifying the two groups to some extent (Figure 1(b)). These results indicated 2.2. Differential Expression Analysis. The count-based expres- that the TFs might be implicated in UC pathogenesis. sion data was used for the differential expression analysis (DEA). R/Bioconductor DESeq2 [15] was employed to iden- 3.2. Expression Patterns of the Differentially Expressed ff tify the di erentially expressed genes (DEGs). The two-fold Transcription Factors. To reveal the expression patterns of p change and adjusted value of 0.05 were used to determine the differentially expressed transcription factors (DETFs), the DEGs for each comparison. we conducted coexpression analysis of the 72 DETFs. Nota- bly, four categories of DETFs (A, B, C, and D) could be iden- fi 2.3. Transcription Factor Target Genes and Pathway ti ed by the coexpression analysis (Figure 2(a)). Further Enrichment Analysis. The Fisher’s exact test was used to iden- analysis of the expression patterns revealed that upregulated TFs in UC (N=RUC the DEGs. The DEGs with a significant correlation with their and C, upregulated TFs in R ( ) had higher pro- portion in group B, and downregulated TFs in UC TFs were selected for this analysis and TFs with a large num- N=R>UC ber of target genes (n > 2000) were excluded in the enrich- ( ) were more frequently observed in group D ment analysis. The enrichment analysis was implemented (Figure 2(b)). These results indicated that three categories in the R clusterProfiler package with enricher function [19]. were observed in these DETFs. 3.3. Target Genes of the DETFs. As the TFs could promote or 2.4. Immune Cell Proportion Estimation. The immune cell suppress the transcription of their target genes, we then proportion was estimated by CIBERSORT, which used the investigated whether the target genes were also differentially gene expression profiles and immune cell-specific genes to expressed. Specifically, DEGs were significantly enriched in characterize the cell composition of complex tissues [20]. the target genes of HOXA2, IKZF1, KLF2, XBP1, EGR2, The count-based expression data was normalized to Tran- ETV7, BACH2, CBFA2T3, HLF, and NFE2 (Figure 3(a), Sup- script Per Million (TPM) by R scater package (https:// plementary Table S3). Remarkably, BACH2, NFE2, IKZF1, bioconductor.riken.jp/packages/3.4/bioc/html/scater.html), EGR2, XBP1, CBFA2T3, and ETV7 were upregulated in UC which was used for the CIBERSORT analysis. (N=RUC or N>R>UC) (Figure 3(b)). It should be noted that BACH2 and NFE2 2.5. Statistical Analysis. The two-sample comparison was were upregulated in UC (Figure 3(c)), and they had t tested by Wilcoxon rank-sum test or test, and multiple- significantly more shared target genes (Figure 3(d)), sample comparison was tested by analysis of variance suggesting that the two TFs might cooperate with each (ANOVA). The Spearman correlation analysis was used to other to regulate their target genes. evaluate the correlation of two variables. Symbols of ∗, ∗∗, ∗∗∗, and ∗∗∗∗ indicate the statistical significances of 0.05, 3.4. Signaling Pathways That the DETFs and Target Genes 0.01, 0.001, and 0.0001, respectively. May Participate in. To further identify the signaling BioMed Research International 3

Remission vs normal UC vs normal UC vs remission

30

15 20 25

20 15 10 15 10

–log10 ( p -value) –log10 ( p -value) 10 –log10 ( p -value) 5 5 5

0 0 0

–1 012 3210–1–2–3 3210–1–2–3 log2 fold change log2 fold change log2 fold change (a)

Group HIF1A 3 PADI4 NFE2 ETV7 STAT1 IRF1 FOXC1 FOXQ1 EOMES TCF7 LEF1 POU2F2 PAX5 LYL1 2 BACH2 ATF3 FOS MSX1 EGR3 EGR2 SPI1 PRDM1 ESR1 ESR2 MYBL1 STAT4 1 LMO2 IKZF1 FLI1 GATA3 ETS1 PRRX2 XBP1 FOXP3 CIITA ARID3A CBFA2T3 TFAP2C 0 KLF2 TP63 NKX3-1 MYBL2 TEAD4 BCL3 FOXP2 PLAG1 HOXA2 HOXA5 HOXA4 NR6A1 –1 NR1H4 PPARG NR1I2 PAX6 FEV SOX10 HLF TEF NR2E3 FOXN1 PAX4 ETV4 –2 NHLH1 TFAP2A PDX1 CYP26A1 PITX2 NANOG FOXD1 FOXI1 KLF1 POU3F1 –3

Group N

R

UC

(b)

Figure 1: Differentially expressed transcription factors (DETFs). (a) The differential expression levels of the TFs. The X and Y axes represent the log2 fold change and -log10 (adjusted p value), respectively. (b) The gene expression profiles of the DETFs in UC, R, and N groups. The red and blue colors represent the high and low expression. 4 BioMed Research International

BACH2 MYBL1 EOMES LMO2 LYL1 PAX5 TCF7 LEF1 GATA3 ETS1 FLI1 IKZF1 STAT4 PRDM1 ESR1 PADI4 ARID3A SPI1 FOXQ1 NFE2 IRF1 ETV7 PRRX2 CBFA2T3 MYBL2 HIF1A XBP1 FOXP3 STAT1 NKX3-1 CIITA POU2F2 TP63 EGR3 EGR2 FOXC1 MSX1 ATF3 FOS KLF1 ESR2 FOXD1 FOXI1 POU3F1 PLAG1 TEAD4 KLF2 BCL3 FOXN1 TFAP2C NHLH1 FOXP2 HLF NR6A1 HOXA2 HOXA5 HOXA4 PAX6 FEV PITX2 NR1I2 NR1H4 PPARG TEF PAX4 PDX1 ETV4 NANOG NR2E3 TFAP2A SOX10 CYP26A1

–0.5 0 0.5 1 Spearman correlation

Clusters A B C D (a)

D

C

B

A

0.0 0.2 0.4 0.6 0.8 1.0 Proportion of genes with different expression patterns

NR=UC NUC N>R>UC NUC N>R

Figure 2: Gene expression patterns of the DETFs. (a) The coexpression modules of the DETFs. The modules were identified by the hierarchical clustering analysis with four clusters. (b) The proportion of DETFs in the eight gene expression patterns. pathways regulated by the DETFs and target genes, we con- were significantly enriched in the pathways (Figure 4(a)). ducted a gene set enrichment analysis of the differentially The virus infection pathways including human papillomavi- expressed target genes of DETFs. We found that target genes rus infection, Epstein-Barr virus infection, Hepatitis B, of BACH2, CBFA2T3, EGR2, ETV7, IKZF1, NFE2, and XBP1 Kaposi sarcoma-associated herpesvirus infection, human BioMed Research International 5

XBP1 TF target gene enrichment p.adjust

NFE2 CBFA2T3

HLF ETV7 NUC BACH2 HOXA2 N>R>UC ETV7 0.008 BACH2 EGR2 N=R

0.016 KLF2 HOXA2

0.010 0.015 0.020 0.025 GeneRatio

Count 10 40

20 50

30 (a) (b) BACH2 NFE2 KCNA2, PEA15, SPATS2, HS3ST3B1, ⁎⁎⁎⁎ ⁎⁎⁎⁎ PPP1R9B, SLC16A6, COL5A3, LINC00152, MMP9, KBTBD8, and 5.5 PRDM1 9.0

8.0 4.5

7.0 3.5 log2 normalized count log2 normalized count

6.0 2.5 NRUC NRUC BACH2 target genes NFE2 target genes Group Group n = 46 n = 53 (c) (d)

Figure 3: The DETFs with significant consequences of their target genes. (a) The DETFs significantly enriched by the DEGs. The node color represents the statistical significance calculated by Fisher’s exact test. The node size represents the number of target genes with differential expression. (b) The expression patterns of nine DETFs significantly enriched by the DEGs. (c) The expression levels of BACH2 and NFE2 in the three groups. (d) The shared target genes between BACH2 and NFE2. immunodeficiency virus 1 infection, and immune-related Particularly, TFs including BACH2, CBFA2T3, EGR2, pathways such as downstream signaling in naïve CD8+ T ETV7, NFE2, and XBP1, and their target genes were signifi- cells and signaling by interleukins were significantly enriched cantly enriched in signaling by interleukins. The inflamma- by these target genes (Figure 4(a)). tory factors such as IL6, IL18RAP, IL11, STAT5B, and CSF3 6 BioMed Research International

p.adjust

Reactome_hemostasis

KEGG_pathways in cancer

Reactome_signaling by receptor tyrosine kinases

Reactome_signaling by interleukins 0.01

KEGG_human papillomavirus infection

Reactome_non-genomic estrogen signaling

Reactome_ESR-mediated signaling 0.02 Reactome_signaling by nuclear receptors

KEGG_Epstein-Barr virus infection

KEGG_hepatitis B

KEGG_endocrine resistance 0.03

KEGG_proteoglycans in cancer

KEGG_Kaposi sarcoma-associated herpesvirus infection

NCI_downstream signaling in naïve CD8+ T cells 0.04 KEGG_MAPK signaling pathway

KEGG_human immunodeficiency virus 1 infection XBP1 NFE2 EGR2 ETV7 IKZF1 BACH2 CBFA2T3

GeneRatio 0.15 0.25 0.20 0.30 (a)

LCP1 AKT3 STAT4 HSP90B1 IL10 STAT3 PTPN6 ETV7 MAP2K1 LYN XBP1 GNGT2 IL6 PSMC2 PSMA1 PIM1 DUSP7 BACH2 NFE2 IL23A MMP7 IL18RAP PSMA3

STAT2 IL13 MMP9 MMP9 CBFA2T3

CBL BATF FASLG ESR-mediated signaling Non-genomic estrogen signaling BACH2 IL11 CSF3 BCL6 EGR2

STAT5B MAP3K3 PTPN7

Signaling by interleukins

Transcription factors

Target genes (b)

Figure 4: The signaling pathways enriched by the target genes of DETFs with differential expression. (a) The DETFs and their regulating signaling pathways. (b) The DETF-target pairs in signaling by interleukins, ESR-mediated signaling, and nongenomic estrogen signaling. were involved in the signaling by interleukins (Figure 4(b)). 3.5. Immune Cells and Their Association with DETFs. As the Furthermore, target genes of BACH2, including AKT3, inflammatory factors and pathways were potentially involved GNGT2, MMP7, and MMP9, were involved in ESR- in UC pathogenesis, we investigated the relative abundance mediated signaling and nongenomic estrogen signaling. of immune cells in mucosal biopsies and their association These results indicated that estrogen signaling and signaling with DETFs. The proportion of immune cells was estimated by interleukins might be closely associated with the UC by CIBERSORT based on the gene expression profiles. Spe- pathogenesis. cifically, proportions of naïve CD4, regulatory T cells (Tregs), BioMed Research International 7

CD4 naive CD8 effector Treg cell ⁎⁎⁎ ⁎ ⁎

0.50 0.08 0.15

0.46 0.06 0.10 0.04 0.42 Proportion (%) Proportion (%) Proportion (%) 0.05 0.02

0.38 0.00 0.00

N R UC N R UC N R UC

Group Group Group

DC pDC Plasma ⁎ ⁎⁎⁎⁎ ⁎⁎ 0.20 0.15

0.24 0.15 0.10 0.20 0.10

0.05 Proportion (%) 0.16 Proportion (%) Proportion (%) 0.05

0.12 0.00 N R UC N R UC N R UC

Group Group Group (a)

1 –0.46 –0.23 –0.21 0.16 0.20 0.35 ETV7

–0.52 –0.40 –0.36 0.13 0.28 0.39 NFE2 0.5 –0.50 –0.30 –0.32 0.03 0.22 0.39 CBFA2T3

–0.53 –0.33 –0.29 0.06 0.30 0.40 BACH2 0

–0.67 –0.44 –0.39 0.17 0.35 0.56 IKZF1

–0.5 –0.61 –0.24 –0.27 0.07 0.31 0.49 EGR2

–0.53 –0.29 –0.43 –0.04 0.30 0.53 XBP1 –1 DC pDC Plasma Treg cell CD4 naive CD8 effector (b)

Figure 5: The association of transcription factors (TFs) with immune cell proportions. (a) The proportion of immune cells that were significantly accumulated or reduced in UC. (b) The Spearman correlation of TFs and immune cell proportions. The red and blue colors represent the positive and negative correlation. 8 BioMed Research International and plasmacytoid dendritic cells (pDC) were decreased in tion of plasma in UC might be associated with increased UC, while effector CD8 and plasma were increased in UC gut permeability [36]. compared with R and N groups (Figure 5(a)). Particularly, In summary, we present a systematic study of the TFs by DC was found to be reduced in the R group (Figure 5(a)). analyzing the DETFs, their regulating target genes and path- The correlation analysis revealed that the nine DETFs with ways, and immune cells. These findings might improve our functional enrichment of pathways including BACH2, understanding of the TFs in the pathogenesis of UC. CBFA2T3, EGR2, ETV7, NFE2, and XBP1 were highly corre- ff lated with e ector CD8 and plasma (Figure 5(b)), indicating Data Availability that these TFs might promote the infiltration of effector CD8 and plasma into the intestinal mucosal tissues. The gene expression data were collected from the Gene Expression Omnibus (GEO) database with accession 4. Discussion GSE128682.

Transcription factors (TFs) are key proteins involved in reg- Conflicts of Interest ulating gene transcription in cells. However, the functional roles of TFs and their regulating target genes and pathways The authors declare that they have no conflicts of interest. are still little known in ulcerative colitis (UC). In the present study, we collected gene expression data of mucosal biopsies from 14 UC, 14 remission (R), and 16 Acknowledgments healthy controls (N), and identified DEGs in the three This manuscript is funded by National Natural Science groups, of which, 72 were identified as differentially Foundation of China (No. 81904152). expressed TFs (DETFs). Furthermore, the coexpression anal- ysis of the DETFs revealed three categories of TFs, which were upregulated in UC (N=RUC), and downregulated in UC (N=R>UC). Supplementary 1. . Supplementary Table S1: UC had signifi- As the function of DETFs could result in dysregulation of ff fi their target genes, we found that DEGs were significantly cantly di erent expression pro les as compared with R and enriched in the target genes of HOXA2, IKZF1, KLF2, N groups. XBP1, EGR2, ETV7, BACH2, CBFA2T3, HLF, and NFE2.As Supplementary 2. . Supplementary Table S2: 72 TFs signifi- BACH2 and NFE2 proteins had similar protein structure cantly differentially expressed between the three groups. [21], they had a greater number of shared target genes. Supplementary 3. . Supplementary Table S3: DEGs were sig- BACH2 has interactions with NFE2L1 and NFE2L3 based nificantly enriched in the target genes. on BIOGRID [22] protein-protein interaction (PPI), indicat- ing that BACH2 might also have the potential to interact with References NFE2. Both BACH2 and NFE2 were implicated in UC via fl regulating in ammation-related pathways [23, 24]. [1] M. Fumery, D. Duricova, C. Gower-Rousseau, V. Annese, Among the TF target genes, inflammatory factors such as L. Peyrin-Biroulet, and P. L. Lakatos, “Review article: the nat- IL6, IL18RAP, IL11, STAT5B, and CSF3 were involved in the ural history of paediatric-onset ulcerative colitis in population- signaling by interleukins. The interleukins and receptors based studies,” Alimentary Pharmacology & Therapeutics, were frequently reported to promote the inflammatory phe- vol. 43, no. 3, pp. 346–355, 2016. notype in UC [25–28]. Notably, IL11 and IL18RAP were [2] Y. Yokoyama, T. Yamakawa, T. Hirano et al., “Current diag- identified as susceptibility loci in UC [29, 30]. Furthermore, nostic and therapeutic approaches to cytomegalovirus infec- target genes of BACH2, including AKT3, GNGT2, MMP7, tions in ulcerative colitis patients based on clinical and basic and MMP9, were involved in ESR-mediated signaling and research data,” International Journal of Molecular Sciences, nongenomic estrogen signaling. As patients with UC have a vol. 21, no. 7, p. 2438, 2020. “ fl higher risk for colorectal carcinoma (CRC) development [3] R. M. Nanau and M. G. Neuman, Metabolome and in amma- fl ” [31] and the estrogen receptors (ER) alpha/beta balance has some in in ammatory bowel disease, Translational Research, fl vol. 160, no. 1, pp. 1–28, 2012. a relevant in uence on colorectal carcinogenesis [32], we “ then speculated that the dysregulation of estrogen signaling [4] T. Tanaka, T. Kobunai, Y. Yamamoto et al., Increased copy might be associated with the risk of colorectal carcinogenesis. number variation of mtDNA in an array-based digital PCR assay predicts ulcerative colitis-associated colorectal cancer,” Furthermore, to clarify the functional roles of immune In Vivo, vol. 31, no. 4, pp. 713–718, 2017. cells on the UC pathogenesis, we estimated the immune cell “ ff [5] K. Kang, J. H. Bae, K. Han, E. S. Kim, Kim TO, and J. M. Yi, A proportions in all the samples. The accumulated e ector genome-wide methylation approach identifies a new hyper- CD8 and reduced proportion of naïve CD4 might be respon- methylated gene panel in ulcerative colitis,” International Jour- sible for the adaptive immune response in UC, showing con- nal of Molecular Sciences, vol. 17, no. 8, p. 1291, 2016. sistency with the previous study [33]. Notably, BACH2 and [6] V. Lorén, A. Garcia-Jaraquemada, J. E. Naves et al., “ANP32E, EGR2 could regulate CD8 cell differentiation, indicating that a protein involved in steroid-refractoriness in ulcerative colitis, + the high proportion of CD8 might be associated with the identified by a systems biology approach,” Journal of Crohn's upregulation of BACH2 and EGR2 [34, 35]. The accumula- & Colitis, vol. 13, no. 3, pp. 351–361, 2019. BioMed Research International 9

[7] K. Dubois-Camacho, D. Diaz-Jimenez, M. de la Fuente et al., [23] K. Christodoulou, A. E. Wiskin, J. Gibson et al., “Next genera- “Inhibition of miR-378a-3p by inflammation enhances IL-33 tion exome sequencing of paediatric inflammatory bowel dis- levels: a novel mechanism of alarmin modulation in ulcerative ease patients identifies rare and novel variants in candidate colitis,” Frontiers in Immunology, vol. 10, p. 2449, 2019. genes,” Gut, vol. 62, no. 7, pp. 977–984, 2013. [8] R. van der Lee, S. Correard, and W. W. Wasserman, “Deregu- [24] Y. D. Jeon, J. H. Lee, Y. M. Lee, and D. K. Kim, “Puerarin lated regulators: disease-causing cis variants in transcription inhibits inflammation and oxidative stress in dextran sulfate factor genes,” Trends in Genetics, vol. 36, no. 7, pp. 523–539, sodium-induced colitis mice model,” Biomedicine & Pharma- 2020. cotherapy, vol. 124, article 109847, 2020. [9] A. Chen and A. N. Koehler, “Transcription factor inhibition: [25] Y. Yan, N. Zhao, X. He, H. Guo, Z. Zhang, and T. Liu, “Mesen- lessons learned and emerging targets,” Trends in Molecular chymal stem cell expression of interleukin-35 protects against Medicine, vol. 26, no. 5, pp. 508–518, 2020. ulcerative colitis by suppressing mucosal immune responses,” – [10] Y. Wen, J. Niu, F. Zhang et al., “Heat shock transcription factor Cytotherapy, vol. 20, no. 7, pp. 911 918, 2018. 2 predicts mucosal healing and promotes mucosal repair of [26] C. A. Parisinos, S. Serghiou, M. Katsoulis et al., “Variation in ulcerative colitis,” Scandinavian Journal of Gastroenterology, interleukin 6 receptor gene associates with risk of Crohn's dis- vol. 55, no. 6, pp. 677–686, 2020. ease and ulcerative colitis,” Gastroenterology, vol. 155, no. 2, – [11] W. Wang, F. Zhang, X. Li et al., “Heat shock transcription fac- pp. 303 306.e2, 2018, e302. tor 2 inhibits intestinal epithelial cell apoptosis through the [27] M. Matusiewicz, K. Neubauer, I. Bednarz-Misa, S. Gorska, and mitochondrial pathway in ulcerative colitis,” Biochemical and M. Krzystek-Korpacka, “Systemic interleukin-9 in inflamma- Biophysical Research Communications, vol. 527, no. 1, tory bowel disease: association with mucosal healing in ulcer- pp. 173–179, 2020. ative colitis,” World Journal of Gastroenterology, vol. 23, – [12] R. K. Weersma, L. Zhou, I. M. Nolte et al., “Runt-related tran- no. 22, pp. 4039 4046, 2017. scription factor 3 is associated with ulcerative colitis and shows [28] I. A. Sroufe, T. Gardner, K. A. Bresnahan, S. M. Quarnberg, epistasis with solute carrier family 22, members 4 and 5,” and P. R. Wiedmeier, “Insights into the pathophysiology of Inflammatory Bowel Diseases, vol. 14, no. 12, pp. 1615–1622, ulcerative colitis: interleukin-13 modulates STAT6 and p38 2008. MAPK activity in the colon epithelial sodium channel,” The [13] C. Guo, F. Yao, K. Wu, L. Yang, X. Zhang, and J. Ding, “Chro- Journal of Physiology, vol. 595, no. 2, pp. 421-422, 2017. matin immunoprecipitation and association study revealed a [29] A. Zhernakova, E. M. Festen, L. Franke et al., “Genetic analysis possible role of runt-related transcription factor 3 in the ulcer- of innate immunity in Crohn's disease and ulcerative colitis ative colitis of Chinese population,” Clinical Immunology, identifies two susceptibility loci harboring CARD9 and vol. 135, no. 3, pp. 483–489, 2010. IL18RAP,” American Journal of Human Genetics, vol. 82, – [14] C. G. Fenton, H. Taman, J. Florholmen, S. W. Sorbye, and no. 5, pp. 1202 1210, 2008. R. H. Paulssen, “Transcriptional signatures that define ulcera- [30] W. Klein, A. Tromm, T. Griga et al., “A polymorphism in the tive colitis in remission,” Inflammatory Bowel Diseases, 2020. IL11 gene is associated with ulcerative colitis,” Genes and – [15] M. I. Love, W. Huber, and S. Anders, “Moderated estimation Immunity, vol. 3, no. 8, pp. 494 496, 2002. of fold change and dispersion for RNA-seq data with DESeq2,” [31] M. Xue, L. Shi, W. Wang, S. Chen, and L. Wang, “An overview Genome Biology, vol. 15, no. 12, article 550, 2014. of molecular profiles in ulcerative colitis-related cancer,” fl – [16] O. Fornes, J. A. Castro-Mondragon, A. Khan et al., “JASPAR In ammatory Bowel Diseases, vol. 24, no. 9, pp. 1883 1894, 2020: update of the open-access database of transcription fac- 2018. tor binding profiles,” Nucleic Acids Research, vol. 48, no. D1, [32] M. Principi, M. Barone, M. Pricci et al., “Ulcerative colitis: pp. D87–D92, 2020. from inflammation to cancer. Do estrogen receptors have a ” [17] E. Wingender, “The TRANSFAC project as an example of role?, World Journal of Gastroenterology, vol. 20, no. 33, – framework technology that supports the analysis of genomic pp. 11496 11504, 2014. regulation,” Briefings in Bioinformatics, vol. 9, no. 4, pp. 326– [33] H. Rabe, M. Malmquist, C. Barkman et al., “Distinct patterns 332, 2008. of naive, activated and memory T and B cells in blood of ” [18] A. Lachmann, H. Xu, J. Krishnan, S. I. Berger, A. R. Mazloom, patients with ulcerative colitis or Crohn's disease, Clinical – and A. Ma'ayan, “ChEA: transcription factor regulation and Experimental Immunology, vol. 197, no. 1, pp. 111 129, inferred from integrating genome-wide ChIP-X experiments,” 2019. Bioinformatics, vol. 26, no. 19, pp. 2438–2444, 2010. [34] R. Roychoudhuri, D. Clever, P. Li et al., “BACH2 regulates ff [19] G. Yu, L. G. Wang, Y. Han, and Q. Y. He, “clusterProfiler: an R CD8(+) T cell di erentiation by controlling access of AP-1 fac- ” – package for comparing biological themes among gene clus- tors to enhancers, Nature Immunology, vol. 17, no. 7, pp. 851 ters,” OMICS, vol. 16, no. 5, pp. 284–287, 2012. 860, 2016. “ [20] A. M. Newman, C. L. Liu, M. R. Green et al., “Robust enumer- [35] N. Du, H. Kwon, P. Li et al., EGR2 is critical for peripheral ff fl ation of cell subsets from tissue expression profiles,” Nature naïve T-cell di erentiation and the T-cell response to in u- ” Methods, vol. 12, no. 5, pp. 453–457, 2015. enza, Proceedings of the National Academy of Sciences of the United States of America, vol. 111, no. 46, pp. 16484–16489, [21] F. Katsuoka and M. Yamamoto, “ proteins (MafF, 2014. MafG, MafK): history, structure and function,” Gene, “ α vol. 586, no. 2, pp. 197–205, 2016. [36] P. P. Trivedi and G. B. Jena, Role of -lipoic acid in dextran sulfate sodium-induced ulcerative colitis in mice: studies on [22] R. Oughtred, C. Stark, B. J. Breitkreutz et al., “The BioGRID inflammation, oxidative stress, DNA damage and fibrosis,” interaction database: 2019 update,” Nucleic Acids Research, Food and Chemical Toxicology, vol. 59, pp. 339–355, 2013. vol. 47, no. D1, pp. D529–D541, 2019.