Intestinal Microbiota and miRNA in IBD: History
Please note this is an old version of this entry, which may differ significantly from the current revision.

Inflammatory bowel disease (IBD) includes Crohn’s disease (CD) and ulcerative colitis (UC) and comprises a chronic gastrointestinal tract disorder characterized by hyperactive and dysregulated immune responses to environmental factors, including gut microbiota and dietary components.

  • intestinal microbiota
  • microRNA
  • dysbiosis
  • inflammatory bowel disease

1. Introduction

Inflammatory bowel disease (IBD) comprises a chronic gastrointestinal tract disorder characterized by hyperactive and dysregulated autoimmune responses and increased intestinal permeability related to environmental factors, including gut microbiota and dietary components, which includes Crohn’s disease (CD) and ulcerative colitis (UC), which differ in their pathophysiological and clinical characteristics [1][2].
The current incidence of CD has increased by 11% compared to the incidence three decades ago [3][4]. The etiology is unknown, and genetic, immunological, and environmental factors contribute to the risk of disease onset and progression. A cure remains elusive, and the efficient management of CD requires a multidisciplinary and interprofessional approach [4][5]. The disease can extend to all segments of the gastrointestinal tract, most commonly the terminal ileum and colon. Inflammation is typically segmental, asymmetrical, and transmural, but over time complications such as strictures, fistulas, or abscesses will develop in half of the patients who often require surgery [4][6][7]. Patients with CD frequently suffer from malnutrition and psychological issues and may have to live with a stoma, which could cause significant morbidity and impact the patients’ quality of life [4][7]. Current therapeutic strategies aim to prevent disease-related complications and interrupt the disease’s recurrence process by prolonging the remission period, whereas personalized medicine and the treat-to-target approach have been the most effective strategies adopted for the control of the inflammatory process [4][7].
UC is a chronic inflammatory condition that causes inflammation of the colon, manifested by continuous lesions and superficial inflammation, which can lead to erosions, ulcers, and bloody diarrhea [8]. The disease is characterized by a relapsing and remitting course, and curative medical therapy is not yet available. Patients with mild or moderate activity are usually unremarkable, apart from blood on rectal examination, whereas patients with a severe attack may exhibit fever, tachycardia, weight loss, abdominal tenderness, abdominal distension, and reduced bowel sounds [8][9].
The management of CD and UC has evolved from the mere treatment of symptoms and induction of clinical remission to more stringent outcomes, including the maintenance of steroid-free remission, the reduction in the number of hospitalizations and surgeries, mucosal and histological healing, improvement in patient-reported outcome, such as the patients’ quality of life [7][8][9][10][11], as well as the control of the risk factors associated with the development of colorectal cancer (CRC). The CRC risk is associated with the duration, extent, severity, and persistence of inflammatory activity [8][12], and it is estimated that CRC can account for up to 10% of deaths in patients with IBD [13].
As the etiology of IBD is not fully understood, it is believed that the interaction among genetic, immunological, and environmental factors, such as intestinal microbiota, can trigger the disease. Chronic gut dysbiosis has also been associated with autoimmune diseases such as eczema, asthma, celiac disease, and type-1 diabetes, as well as with diseases related to the consumption of an unbalanced diet, increased inactivity, age, obesity, type 2 diabetes, metabolic and cardiovascular diseases [14][15], liver disorders such as non-alcoholic liver steatosis [16], cancers such as colorectal cancer, and psychological diseases such as depression, anxiety, autism and Alzheimer’s disease [17][18][19].
During inflammation in IBD, oxidative stress promotes an increase in pathogenic bacteria at the expense of beneficial bacteria. This can cause an imbalance in the intestinal microbiota, potentially making it a useful biomarker and predictor for stratifying patients with IBD [20][21][22]. Further, microbiota-modulating therapies such as diet, fecal microbiota transplantation, pre- and probiotics, symbiotics, and antibiotics have been studied as potential therapies, thereby demonstrating the importance of intestinal microbiota in IBD patients [22][23][24][25][26].
miRNAs are potential disease markers that have been studied in recent years. miRNAs have been involved in the pathogenesis of IBD and their role has been studied both as a diagnostic biomarker and also as therapeutic targets [27][28]. miRNAs may represent a useful tool in the differentiation between UC and CD, besides being adopted as biomarkers of disease activity, of response to therapy as well as the potential to be used as prognostic markers of disease severity and the presence of complications such as stenosing, penetrating disease and CRC.
Both the intestinal microbiota and miRNAs have been the target of recent studies aimed at gaining an appreciation of their roles and relationship with IBD [29][30][31][32]. Considering that intestinal microbiota and miRNAs are both strongly related to IBD, understanding the role of each in the IBD process can provide vital information that could result in the development of more effective and accurate diagnostic tools and target treatments for patients with IBD.

2. MicroRNAs in Inflammatory Bowel Disease

miRNAs are a group of small (18–24 nucleotides), single-stranded, non-coding RNA molecules that can act as potent negative regulators in gene expression [33][34]. Each miRNA can target hundreds of mRNAs within a given cell type, and a single mRNA is often the target of multiple miRNAs. Thus, miRNAs contribute to the regulation of >30% of protein-coding genes [35]. Several biological processes are regulated by miRNAs, including cell survival, differentiation, proliferation, apoptosis, cell cycle control, and homeostasis; additionally, specific miRNAs regulate the differentiation of intestinal epithelial cells [33][34].
miRNAs have been extensively studied in multiple types of cancer and have been reported as regulators of tumor suppressors and oncogenes [35]. Although most studies are focused on their aforementioned role, the impact on autoimmune diseases and especially IBD is not fully investigated (Table 1). It has been suggested that the critical function of these small RNAs is to contribute to the establishment of immunological homeostasis at mucosal sites [33].
The first study reporting miRNA alterations in IBD patients revealed 11 different expressed miRNAs in patients with UC vs. controls. The miRNAs miR-16, miR-21, miR-23a, miR-24, miR-29a, miR-126, miR-195, and let-7f were increased, whereas miR-192, miR-375, and miR-422b were reduced [36]. Subsequently, several studies have been conducted with the aim of characterizing such alterations in the expression of miRNAs [37][38][39] and, consequently, several of these miRNAs have been suggested as potential biomarkers for CD or UC both in colonic tissues and non-invasive samples such as blood and feces [39][40][41][42].
The distinct miRNA expression was described in tissues from different intestinal regions in patients with active ileal or colonic CD [37]. Three miRNAs were increased (miR-31, miR-215, miR-22), and one miRNA (miR-19b) was decreased in the terminal ileum compared to those in the colon, supporting the likelihood that miRNAs influence different inflammation-related gene expression in each IBD subtype [37].
Another study reported miR-223 as a potential biomarker in the serum of patients with IBD [42]. Patients with IBD had significantly increased serum levels of miR-223 to controls, showing a positive correlation with disease activity in patients with CD and UC. Moreover, miR-223 showed a better disease activity correlation in patients with CD compared to erythrocyte sedimentation rate and high-sensitivity C-reactive protein [42]. In another study, the circulating miR-320a levels were strongly correlated with endoscopic disease activity in patients with CD and UC, highlighting its potential as a non-invasive biomarker in monitoring the control of the inflammatory process [43].
In addition to their potential role in monitoring disease activity, whether in clinical [42], biochemical [42], or endoscopic activity [43], miRNA can also be used as predictors of response to therapy. When evaluating patients with severe UC, Morilla et al. [44] identified 15 miRNAs associated with the response to corticosteroids, 6 miRNAs associated with the response to infliximab, and 4 associated with the response to cyclosporine, in those patients unresponsive to initial corticosteroid therapy, thereby highlighting the role of miRNA as a predictor of response to therapy in IBD. In other study including children with IBD (CD: 17 and UC: 2) who received prednisone or infliximab, miR-146a, miR-320a, and miR-146b decreased with both drugs, correlating to the control of the inflammatory process, and miR-486 showed a significant change in response to prednisone but not to infliximab [45].
Table 1. The main microRNA involved in Inflammatory Bowel Disease patients.
miRNA In IBD Target References
miR10a Decrease Inhibit NOD2 [46]
miR-16 Increase in UC T-cell sub-types [36]
miR-19b Decrease in CD   [37]
miR-21 Increase in UC T-cell sub-types [36]
miR-22 Increase in CD Th17 cell [37]
miR-23a Increase in UC   [36]
miR-24 Increase in UC   [36]
miR-29 Decrease IL-12 and IL-23 in CD Through activation of NOD2 [47][48]
miR-29a Increase in UC NOD2 [48]
miR-31 Increase in CD   [37]
miR-107 Decrease IL-23p19 (a subunit of IL-23) [49]
miR-126 Increase Regulates VCAM-1 [36][50][51]
miR-143/145 Decrease Inhibit IGFBP5 (regulate IGF pathway in intestinal epithelial regeneration) [32][52]
miR-146a Increase TNF-α [45]
miR-146b Increase TNF-α [45]
miR-150 Increase intestinal permeability c-Myb [32][53]
miR-155 Increase in UC SOCS1 [54]
miR-192 Decrease in UC MIP-2α [36]
miR-195 Increase in UC   [36]
miR-215 Increase in CD MIP-2α [37]
miR-223 Increase Claudin-8 (a TJ-integral protein) [42]
miR-320a Increase   [43][45]
miR-375 Decrease in UC inhibit KLF5 (antagonist of the goblet cell–differentiation factor KLF4) [32][36]
miR-422b Decrease in UC   [36]
miR-486 Increase   [45]
let-7f Increase in UC T-cell sub-types [36]
Relationship of main microRNA with decreased or increased activity in Inflammatory Bowel Disease (IBD). CD: Crohn’s disease; IGFBP5: insulin-like growth factor-binding protein 5; KLF5/4: Kruppel-like factor 5 and 4; MIP-2α: macrophage inhibitory peptide; NOD2: nucleotide-binding oligomerization domain 2; SOCS1: Suppressor of Cytokine Signaling 1; Th17: T helper 17; TJ: tight junction; TNF-α: tumor necrosis factor alpha; UC: ulcerative colitis; VCAM-1: Vascular Cell Adhesion Molecule-1.
In addition to their role as markers of inflammatory activity, miRNAs may be the actual therapeutic target in the future. Some identified miRNAs act on the same inflammatory pathways as some medications approved for the treatment of IBD. miR-29 has been observed to comprise a family of miRNAs with the potential to decrease levels of IL-23 [48][50], such as ustekinumab, an antibody that inhibits IL12/23, indicated in moderate to severe CD. miR-126 inhibits the leukocyte adhesion to endothelial cells through the regulation of Vascular Cell Adhesion Molecule-1 (VCAM-1) [50][51], the same mechanism of action of the vedolizumab, also indicated for the treatment of moderate to severe IBD. The miR-155 Suppressor of Cytokine Signaling 1 (SOCS1) targets a regulatory protein of the Janus Kinase (JAK) signaling pathway [54], mimicking the use of JAK inhibitors currently available for UC treatment. A review published by Moein et al. [55] clarified the relationship among various miRNAs and the mechanisms involved in the pathogenesis of IBD, including modulations of the inflammatory response through dendritic cells, macrophages, neutrophils, natural killer cells and T cells, dysregulation of TJs, formation of the mucous barrier, and regulation of apoptosis. The elucidation of the role of miRNAs in the inflammatory cascade opens new innovative perspectives for the treatment of IBD, for example, by providing the enhancement of miRNAs that act by inhibiting the inflammatory response (using RNA mimics) or inhibiting the miRNAs that act to perpetuate the inflammatory response (using antagonists of miRNAs).
Changes in the profiles of miRNAs could represent a useful tool in the differentiation of UC and CD, providing important information about the pathophysiology of each disease, prognosis, and response to therapy. Moreover, the identification of the dysregulated miRNA may represent new targets for new therapies focusing on modulation of the inflammatory process via miRNA regulation. Future studies are required for better characterization of the miRNA profile in IBD patients and to clarify the role of miRNAs in triggering and maintaining the inflammatory process in IBD patients and their applications in clinical practice.

This entry is adapted from the peer-reviewed paper 10.3390/ijms24087176

References

  1. Hills, R.D.; Pontefract, B.A.; Mishcon, H.R.; Black, C.A.; Sutton, S.C.; Theberge, C.R. Gut microbiome: Profound implications for diet and disease. Nutrients 2019, 11, 1613.
  2. Oligschlaeger, Y.; Yadati, T.; Houben, T.; Condello Oliván, C.M.; Shiri-Sverdlov, R. Inflammatory Bowel Disease: A Stressed “Gut/Feeling”. Cells 2019, 8, 659.
  3. Zhao, M.; Gönczi, L.; Lakatos, P.L.; Burisch, J. The Burden of Inflammatory Bowel Disease in Europe in 2020. J. Crohn’s Colitis 2021, 15, 1573–1587.
  4. Adamina, M.; Bonovas, S.; Raine, T.; Spinelli, A.; Warusavitarne, J.; Armuzzi, A.; Bachmann, O.; Bager, P.; Biancone, L.; Bokemeyer, B.; et al. ECCO Guidelines on Therapeutics in Crohn’s Disease: Surgical Treatment. J. Crohn’s Colitis 2020, 14, 155–168.
  5. Roda, G.; Chien, N.S.; Kotze, P.G.; Argollo, M.; Panaccione, R.; Spinelli, A.; Kaser, A.; Peyrin-Biroulet, L.; Danese, S. Crohn’s disease. Nat. Rev. Dis. Prim. 2020, 17, 22.
  6. Torres, J.; Mehandru, S.; Colombel, J.F.; Peyrin-Biroulet, L. Crohn’s disease. Lancet 2017, 389, 1741–1755.
  7. Torres, J.; Bonovas, S.; Doherty, G.; Kucharzik, T.; Gisbert, J.P.; Raine, T.; Adamina, M.; Armuzzi, A.; Bachmann, O.; Bager, P.; et al. ECCO Guidelines on Therapeutics in Crohn’s Disease: Medical treatment. J. Crohn’s Colitis 2020, 14, 4–22.
  8. Magro, F.; Gionchetti, P.; Eliakim, R.; Ardizzone, S.; Armuzzi, A.; Barreiro-de Acosta, M.; Burisch, J.; Gecse, K.B.; Hart, A.L.; Hindryckx, P.; et al. Third European evidence-based consensus on diagnosis and management of ulcerative colitis. Part 1: Definitions, diagnosis, extra-intestinal manifestations, pregnancy, cancer surveillance, surgery, and ileo-anal pouch disorders. J. Crohn’s Colitis 2017, 11, 649–670.
  9. Harbord, M.; Eliakim, R.; Bettenworth, D.; Karmiris, K.; Katsanos, K.; Kopylov, U.; Kucharzik, T.; Molnár, T.; Raine, T.; Sebastian, S.; et al. Third European evidence-based consensus on diagnosis and management of ulcerative colitis. Part 2: Current management. J. Crohn’s Colitis 2017, 11, 769–784.
  10. Ordás, I.; Eckmann, L.; Talamini, M.; Baumgart, D.C.; Sandborn, W.J. Ulcerative colitis. Lancet 2012, 380, 1606–1619.
  11. Danese, S.; Roda, G.; Peyrin-Biroulet, L. Evolving therapeutic goals in ulcerative colitis: Towards disease clearance. Nat. Rev. Gastroenterol. Hepatol. 2020, 17, 1–2.
  12. Quaglio, A.E.V.; Grillo, T.G.; Oliveira, E.C.S.D.; Stasi, L.C.D.; Sassaki, L.Y. Gut microbiota, inflammatory bowel disease and colorectal cancer. World J. Gastroenterol. 2022, 28, 4053–4060.
  13. Nadeem, M.S.; Kumar, V.; Al-Abbasi, F.A.; Kamal, M.A.; Anwar, F. Risk of colorectal cancer in inflammatory bowel diseases. Semin. Cancer Biol. 2020, 64, 51–60.
  14. Harmsen, H.J.M.; de Goffau, M.C. The Human Gut Microbiota. In Microbiota of the Human Body—Implications in Healthy and Disease, 1st ed.; Schwiertz, A., Ed.; Springer: Cham, Switzerland, 2016; pp. 95–108.
  15. Lynch, S.V.; Pedersen, O. The human intestinal microbiome in health and disease. N. Engl. J. Med. 2016, 375, 2369–2379.
  16. Milosevic, I.; Vujovic, A.; Barac, A.; Djelic, M.; Korac, M.; Spurnic, A.R.; Gmizic, I.; Stevanovic, O.; Djordjevic, V.; Levic, N.; et al. Gut-liver axis, gut microbiota, and its modulation in the management of liver diseases: A review of the literature. Int. J. Mol. Sci. 2019, 20, 395.
  17. Altveş, S.; Yildiz, H.K.; Vural, H.C. Interaction of the microbiota with the human body in health and diseases. Biosci. Microb. Food Health 2020, 39, 23–32.
  18. Treisman, G.J. The Role of the Brain–Gut–Microbiome in Mental Health and Mental Disorders. In The Microbiota in Gastrointestinal Pathophysiology, 1st ed.; Floch, M., Ringel, Y., Walker, W.A., Eds.; Elsevier: Amsterdam, The Netherlands, 2017; pp. 389–397.
  19. Dai, Z.; Coker, O.O.; Nakatsu, G.; Wu, W.K.K.; Zhao, L.; Chen, Z.; Chan, F.K.L.; Kristiansen, K.; Sung, J.J.Y.; Wong, S.H.; et al. Multi-cohort analysis of colorectal cancer metagenome identified altered bacteria across populations and universal bacterial markers. Microbiome 2018, 6, 70.
  20. Zheng, L.; Wen, X.-L. Gut microbiota and inflammatory bowel disease: The current status and perspectives. World. J. Clin. Cases 2021, 9, 321–333.
  21. Guo, X.; Huang, C.; Xu, J.; Xu, H.; Liu, L.; Zhao, H.; Wang, J.; Huang, W.; Peng, W.; Chen, Y.; et al. Gut Microbiota Is a Potential Biomarker in Inflammatory Bowel Disease. Front. Nutr. 2022, 8, 1340.
  22. Zhang, Y.; Li, Y.; Ren, X.; Zhang, X.; Wu, Z.; Liu, L. The positive correlation of antioxidant activity and prebiotic effect about oat phenolic compounds. Food Chem. 2023, 402, 134231.
  23. Zhou, J.; Li, M.; Chen, Q.; Li, X.; Chen, L.; Dong, Z.; Zhu, W.; Yang, Y.; Liu, Z.; Chen, Q. Programmable probiotics modulate inflammation and gut microbiota for inflammatory bowel disease treatment after effective oral delivery. Nat. Commun. 2022, 13, 3432.
  24. Yang, Y.; Zheng, X.; Wang, Y.; Tan, X.; Zou, H.; Feng, S.; Zhang, H.; Zhang, Z.; He, J.; Cui, B.; et al. Human Fecal Microbiota Transplantation Reduces the Susceptibility to Dextran Sulfate Sodium-Induced Germ-Free Mouse Colitis. Front. Immunol. 2022, 13, 836542.
  25. Dixit, K.; Chaudhari, D.; Dhotre, D.; Shouche, Y.; Saroj, S. Restoration of dysbiotic human gut microbiome for homeostasis. Life Sci. 2021, 278, 119622.
  26. Martyniak, A.; Medyńska-Przęczek, A.; Wędrychowicz, A.; Skoczeń, S.; Tomasik, P.J. Prebiotics, probiotics, synbiotics, paraprobiotics and postbiotic compounds in IBD. Biomolecules 2021, 11, 1903.
  27. Feng, Y.; Zhang, Y.; Zhou, D.; Chen, G.; Li, N. MicroRNAs, intestinal inflammatory and tumor. Bioorganic Med. Chem. Lett. 2019, 29, 2051–2058.
  28. James, J.P.; Riis, L.B.; Malham, M.; Høgdall, E.; Langholz, E.; Nielsen, B.S. MicroRNA biomarkers in IBD-differential diagnosis and prediction of colitis-associated cancer. Int. J. Mol. Sci. 2020, 21, 7893.
  29. Johnston, D.G.W.; Williams, M.A.; Thaiss, C.A.; Cabrera-Rubio, R.; Raverdeau, M.; McEntee, C.; Cotter, P.D.; Elinav, E.; O’Neill, L.A.J.; Corr, S.C. Loss of microRNA-21 influences the gut microbiota, causing reduced susceptibility in a murine model of colitis. J. Crohn’s Colitis 2018, 12, 835–848.
  30. Wortelboer, K.; Bakker, G.J.; Winkelmeijer, M.; Van Riel, N.; Levin, E.; Nieuwdorp, M.; Herrema, H.; Davids, M. Fecal microbiota transplantation as tool to study the interrelation between microbiota composition and miRNA expression. Microbiol. Res. 2022, 257, 126972.
  31. Viennois, E.; Chassaing, B.; Tahsin, A.; Pujada, A.; Wang, L.; Gewirtz, A.T.; Merlin, D. Host-derived fecal microRNAs can indicate gut microbiota healthiness and ability to induce inflammation. Theranostics 2019, 9, 4542–4557.
  32. Algieri, F.; Garrido-Mesa, J.; Vezza, T.; Rodríguez-Sojo, M.J.; Rodríguez-Cabezas, M.E.; Olivares, M.; Garcia, F.; Gálvez, J.; Morón, R.; Rodríguez-Nogales, A. Intestinal anti-inflammatory effects of probiotics in DNBS-colitis via modulation of gut microbiota and microRNAs. Eur. J. Nutr. 2020, 60, 2537–2551.
  33. Archanioti, P.; Gazouli, M.; Theodoropoulos, G.; Vaiopoulou, A.; Nikiteas, N. Micro-RNAs as regulators and possible diagnostic bio-markers in inflammatory bowel disease. J. Crohn’s Colitis 2011, 5, 520–524.
  34. Coskun, M.; Bjerrum, J.T.; Seidelin, J.B.; Nielsen, O.H. MicroRNAs in inflammatory bowel disease-pathogenesis, diagnostics and therapeutics. World. J. Gastroenterol. 2012, 18, 4629–4634.
  35. Dalal, S.R.; Kwon, J.H. The role of microRNA in inflammatory bowel disease. Gastroenterol. Hepatol. 2010, 6, 714–722.
  36. Wu, F.; Zikusoka, M.; Trindade, A.; Dassopoulos, T.; Harris, M.L.; Bayless, T.M.; Brant, S.R.; Chakravarti, S.; Kwon, J.H. MicroRNAs Are Differentially Expressed in Ulcerative Colitis and Alter Expression of Macrophage Inflammatory Peptide-2α. Gastroenterology 2008, 135, 1624–1635.
  37. Wu, F.; Zhang, S.; Dassopoulos, T.; Harris, M.L.; Bayless, T.M.; Meltzer, S.J.; Brant, S.R.; Kwon, J.H. Identification of microRNAs associated with ileal and colonic Crohn’s disease. Inflamm. Bowel Dis. 2010, 16, 1729–1738.
  38. Mohammadi, A.; Kelly, O.B.; Filice, M.; Kabakchiev, B.; Smith, M.I.; Silverberg, M.S. Differential expression of microRNAs in peripheral blood mononuclear cells identifies autophagy and TGF-beta-related signatures aberrantly expressed in inflammatory bowel disease. J. Crohn’s Colitis 2018, 12, 568–581.
  39. Masi, L.; Capobianco, I.; Magrì, C.; Marafini, I.; Petito, V.; Scaldaferri, F. MicroRNAs as Innovative Biomarkers for Inflammatory Bowel Disease and Prediction of Colorectal Cancer. Int. J. Mol. Sci. 2022, 23, 7991.
  40. Suri, K.; Bubier, J.A.; Wiles, M.V.; Shultz, L.D.; Amiji, M.M.; Hosur, V. Role of microRNA in inflammatory bowel disease: Clinical evidence and the development of preclinical animal models. Cells 2021, 10, 2204.
  41. Schönauen, K.; Le, N.; Von Arnim, U.; Schulz, C.; Malfertheiner, P.; Link, A. Circulating and fecal microRNAs as biomarkers for inflammatory bowel diseases. Inflamm. Bowel Dis. 2018, 24, 1547–1557.
  42. Wang, H.; Zhang, S.; Yu, Q.; Yang, G.; Guo, J.; Li, M.; Zeng, Z.; He, Y.; Chen, B.; Chen, M. Circulating MicroRNA223 is a new biomarker for inflammatory bowel disease. Medicine 2016, 95, e2703.
  43. Cordes, F.; Demmig, C.; Bokemeyer, A.; Brückner, M.; Lenze, F.; Lenz, P.; Nowacki, T.; Tepasse, P.; Schmidt, H.H.; Schmidt, M.A.; et al. MicroRNA-320a Monitors Intestinal Disease Activity in Patients With Inflammatory Bowel Disease. Clin. Transl. Gastroenterol. 2020, 11, e00134.
  44. Morilla, I.; Uzzan, M.; Laharie, D.; Cazals-Hatem, D.; Denost, Q.; Daniel, F.; Belleannee, G.; Bouhnik, Y.; Wainrib, G.; Panis, Y.; et al. Colonic MicroRNA Profiles, Identified by a Deep Learning Algorithm, That Predict Responses to Therapy of Patients with Acute Severe Ulcerative Colitis. Clin. Gastroenterol. Hepatol. 2019, 17, 905–913.
  45. Heier, C.R.; Fiorillo, A.A.; Chaisson, E.; Gordish-Dressman, H.; Hathout, Y.; Damsker, J.M.; Hoffman, E.P.; Conklin, L.S. Identification of Pathway-Specific Serum Biomarkers of Response to Glucocorticoid and Infliximab Treatment in Children with Inflammatory Bowel Disease. Clin. Transl. Gastroenterol. 2016, 7, e192.
  46. Zhou, H.; Xiao, J.; Wu, N.; Liu, C.; Xu, J.; Liu, F.; Wu, L. MicroRNA-223 Regulates the Differentiation and Function of Intestinal Dendritic Cells and Macrophages by Targeting C/EBPβ. Cell Rep. 2015, 13, 1149–1160.
  47. Bae, H.J.; Noh, J.H.; Kim, J.K.; Eun, J.W.; Jung, K.H.; Kim, M.G.; Chang, Y.G.; Shen, Q.; Kim, S.J.; Park, W.S.; et al. MicroRNA-29c functions as a tumor suppressor by direct targeting oncogenic SIRT1 in hepatocellular carcinoma. Oncogene 2014, 33, 2557–2567.
  48. Chapman, C.G.; Pekow, J. The emerging role of miRNAs in inflammatory bowel disease: A review. Therap. Adv. Gastroenterol. 2015, 8, 4–22.
  49. Chivukula, R.R.; Shi, G.; Acharya, A.; Mills, E.W.; Zeitels, L.R.; Anandam, J.L.; Abdelnaby, A.A.; Balch, G.C.; Mansour, J.C.; Yopp, A.C.; et al. An essential mesenchymal function for miR-143/145 in intestinal epithelial regeneration. Cell 2014, 157, 1104–1116.
  50. Harris, T.A.; Yamakuchi, M.; Ferlito, M.; Mendell, J.T.; Lowenstein, C.J. MicroRNA-126 regulates endothelial expression of vascular cell adhesion molecule 1. Proc. Natl. Acad. Sci. USA 2008, 105, 1516–1521.
  51. Fedyk, E.R.; Wyant, T.; Yang, L.L.; Csizmadia, V.; Burke, K.; Yang, H.; Kadambi, V.J. Exclusive antagonism of the α4β7 integrin by vedolizumab confirms the gut-selectivity of this pathway in primates. Inflamm. Bowel Dis. 2012, 18, 2107–2119.
  52. Feng, Q.; Li, Y.; Zhang, H.; Wang, Z.; Nie, X.; Yao, D.; Han, L.; Chen, W.D.; Wang, Y.D. Deficiency of miRNA-149-3p shaped gut microbiota and enhanced dextran sulfate sodium-induced colitis. Mol. Ther.-Nucleic Acids 2022, 30, 208–225.
  53. Casado-Bedmar, M.; Viennois, E. MicroRNA and Gut Microbiota: Tiny but Mighty-Novel Insights into Their Cross-talk in Inflammatory Bowel Disease Pathogenesis and Therapeutics. J. Crohns Colitis 2022, 16, 992–1005.
  54. Pathak, S.; Grillo, A.R.; Scarpa, M.; Brun, P.; D’Incà, R.; Nai, L.; Banerjee, A.; Cavallo, D.; Barzon, L.; Palú, G.; et al. MiR-155 modulates the inflammatory phenotype of intestinal myofibroblasts by targeting SOCS1 in ulcerative colitis. Exp. Mol. Med. 2015, 47, e164.
  55. Moein, S.; Vaghari-Tabari, M.; Qujeq, D.; Majidinia, M.; Nabavi, S.M.; Yousefi, B. MiRNAs and inflammatory bowel disease: An interesting new story. J. Cell Physiol. 2019, 234, 3277–3293.
More
This entry is offline, you can click here to edit this entry!
Video Production Service