Loading

Commentary Open Access
Volume 3 | Issue 5 | DOI: https://doi.org/10.33696/immunology.3.116

Immune Cells and Transcriptional Signatures Revealed Novel Regulators and Predict Clinical Response to Biologic Therapy in Ulcerative Colitis

  • 1Department of Pathology and Laboratory Medicine, Tulane University; 1430 Tulane Ave SL-79; New Orleans, LA 70112 USA
+ Affiliations - Affiliations

*Corresponding Author

Suzana D. Savkovic,  ssavkovi@tulane.edu

Received Date: August 06, 2021

Accepted Date: October 13, 2021

Commentary

Inflammatory Bowel Disease (IBD), which includes Crohn’s Disease (CD) and Ulcerative Colitis (UC), has a heterogeneous pathogenesis underlined by genetic predisposition, intestinal barrier dysfunction, impaired immune response, and microbiota imbalance [1-3]. This proceeds to aberrant immune cells presence and function in the affected tissue, activation of signaling pathways, and expression of regulators that subsequently drive inflammation [2,4-7]. Using publicly available transcriptomes obtained from large number of UC patients from European and the US cohorts [8-16], we identified systemic immune cell landscape, pathways, and transcriptional signatures specific for UC as well as those determining outcome of biologic therapy [17].

The global immune cell landscape in UC colonic tissue, determined by CIBERSORT [18] assessment of transcriptomes, revealed systemically elevated levels of neutrophils, T CD4 memory activated cells, active dendritic cells (DC), M0/M1 macrophages, and B naïve cells. Several cell subsets were noticeably reduced such as T CD8, Tregs, B memory, and M2 macrophages. In addition, both resting DC and resting mast cells were lowered, while their active forms were elevated in UC tissue [17]. Relative abundances of other cell subsets in UC tissue were also altered but did not meet significant threshold with applied criteria. Individually, these immune cells are recognized to have important roles in intestinal inflammation [4-6]. Our findings demonstrated that this systemic immune cell landscape was common across UC patients in multiple cohorts. Elevated level of neutrophils in intestinal tissue, an early sign of inflammation, fuels disease by augmenting tissue damage [6,19]. Aberrant T cells and macrophages in the intestine are critical for facilitating inflammatory responses and injury [4,5,20,21]. Some immune cells have dual functions, such as B cells and DC, which are initially protective, but in the long run contribute to UC pathobiology [22-24]. Further, immune cells including macrophages can foster each other’s activity and stimulate other immune cells in promoting inflammation [5,25-27]. Moreover, decrease in certain cells in the intestine impairs protection from bacterial products from the lumen and weakens antigen presentation and processing [28]. Furthermore, in uninflamed (matched) colonic tissue from patients with active UC the majority of the samples displayed an immune cell landscape similar to healthy colonic tissue. However, presence of certain cells (CD4 memory activated and Tregs) was similar to inflamed UC tissue, while presence of other cells (subsets of DC and macrophages) differed from both healthy and inflamed UC. Thus, uninflamed colonic tissue from patients with active UC may provide important information about pathogenesis and recurrent inflammatory episodes. Additionally, we found that a subset of T CD4 cells differed between cohorts, suggesting that these cells may be responsible for disease relapse [21,29]. We speculate that this difference between patient cohorts could be due to variations in composition of microbiota and diversity of regional diet. Similarly, differences in eosinophils, which are involved in protecting intestinal barrier integrity and immunity, might be related to geographic and seasonal disparities among UC cohorts [30,31]. Furthermore, UC tissue obtained from patients prior to biologic therapy with anti-TNFα and anti-α4β7, which were later identified clinically as non-responders, had considerably more neutrophils and T CD4 activated cells when compared to responders. Clinical studies demonstrated that non-responsiveness to the biologic therapy is, in part, related to disease severity, patients age at diagnosis, and duration of inflammation [32-35]. Similar findings are recently reported using single-cell sequencing of a UC cohort [36]. The single-cell sequencing approach provides insight into the existence and behavior of different cell types, while CIBERSORT allows analysis of transcriptomes from multiple cohorts as well as from already existing transcriptomic data. This supports the importance of different approaches to understand the complexity of cell composition in UC pathobiology.

Using Ingenuity Pathway Analysis, we discovered signaling pathways associated with differentially expressed genes (DEGs) in UC tissue compared to controls across different cohorts [17]. These systemic pathways are linked to bacterial response, inflammation, and intracellular signaling. Further, we identified a transcriptional signature consisting of the top hundred DEGs (UC100) that are common across three different cohorts [17]. In an independent cohort, the UC100 distinctly separated inflamed from non-inflamed samples via hierarchical clustering. Among the DEGs in the UC100 are those with established roles in UC pathobiology including regulators of hypoxia, nitric oxide, inflammation, matrix metallopeptidases, and calcium signaling [6,37-40]. Moreover, we selected DEGs whose role in IBD are not well examined for validation in primary UC tissue (by qPCR) that encode regulators of lipid metabolism and mitochondrial function. As for regulators of lipid metabolism, we found increased LPCAT1, which controls lipid droplet number and size [41], LIPG, involved in lipoprotein metabolism [42], and HCAR3, which regulates lipolysis [43,44]. Increased lipid metabolism and intracellular lipid droplets drive intestinal inflammation [45-47]. In addition, LIPG may be involved in endothelial biology [42] and HCAR3 may play roles in crosstalk between metabolites derived from microbiota and immune cells [43,44]. Moreover, DEGs encoding regulators of mitochondrial function including ACAT1 and HMGCS2 are decreased in UC tissues. Limited studies demonstrated that loss of HMGCS2 function in intestinal stem cells could impact intestinal barrier renewal and function [48,49], while ACAT1 has been recently implicated in inflammatory responses in macrophages [50]. In IBD, aberrant expression of mitochondrial regulators leads to reduced respiratory activity, which may further exacerbate response to bacterial signaling [51-55]. A recent study revealed that mitochondrial fission-fusion is critical in homeostasis of intestinal cells and macrophages [56]. Mitochondrial reprograming may also depend on environmental factors, such as use of antibiotics and intake of a high-fat western diet [57]. The exact mechanisms and role of metabolic reprograming with lipids and mitochondria in intestinal cells and immune cells are not fully understood. Their emergence as a hallmark of intestinal inflammation highlights the importance of lipids and mitochondria in the underlying mechanism of disease.

Furthermore, twenty “resistant” DEGs (UC20R) from transcriptomes of UC patients that were common for non-responders to both anti-TNFα and anti-α4β7 therapy were identified [17]. DEGs within UC20R encode regulators involved in bacteria response, defense response, cell signaling, cell trafficking, endothelial function, and metabolism. The UC20R transcriptional signature had significant predictive power for determining (non) response to both anti-TNFα and anti-α4β7 therapy as demonstrated by receiver operating characteristic (ROC) curve analysis and calculating area under the curve (AUC) [17]. Several DEGs with the highest prediction (sensitivity of 73.3% and specificity 85.7%) for non-responding to both therapies, including SELE, VNN2, and STC1, have critical roles in neutrophil accumulation and transendothelial movement at sites of inflammation [58]. This suggests a possible role of immune cell trafficking in predicting response to biologic therapy. Moreover, these features of disease could also be, in part, mediated by changes in the microbiota caused by therapy [59]. Therefore, identifying additional resistant signatures in UC patients may provide guidance for using select therapy and more personalized therapeutic approaches.

A comprehensive assessment of transcriptomes from UC patient colonic tissue demonstrated shared and distinct immune cell landscape, signaling pathways, and transcriptional signatures among cohorts. Further development of new approaches differentiating active from non-active immune cells and interactive from noninteractive cells may provide a platform for more precisionbased identifiers of cell heterogenicity in affected tissue. Additionally, combination of bioinformatics approach with human genetics, epigenetics, and single-cell genomics will lead to mechanistic understanding of inflammatory disorders, risks of recurrence, and association with treatment outcomes. Consequently, it is plausible that these directions may provide clues for development of more precise, personalized diagnostics and therapeutic intervention for UC.

 

References

0.

1. Ponder A, Long MD. A clinical review of recent findings in the epidemiology of inflammatory bowel disease. Clin Epidemiol. 2013;5:237-47.

2. Abreu MT. The genetics and pathogenesis of inflammatory bowel disease. Gastroenterol Hepatol (N Y).2013;9(8):521-3.

3. Liu TC, Stappenbeck TS. Genetics and Pathogenesis of Inflammatory Bowel Disease. Annu Rev Pathol.2016;11:127-48.

4. Okumura R, Takeda K. Maintenance of gut homeostasis by the mucosal immune system. Proc Jpn Acad Ser B Phys Biol Sci. 2016;92(9):423-35.

5. Hisamatsu T, Kanai T, Mikami Y, Yoneno K, Matsuoka K, Hibi T. Immune aspects of the pathogenesis of inflammatory bowel disease. Pharmacology & Therapeutics. 2013;137(3):283-97.

6. de Souza HS, Fiocchi C. Immunopathogenesis of IBD: current state of the art. Nat Rev Gastroenterol Hepatol. 2016;13(1):13-27.

7. Charo IF, Ransohoff RM. The many roles of chemokines and chemokine receptors in inflammation. N Engl J Med. 2006;354(6):610-21.

8. Olsen J, Gerds TA, Seidelin JB, Csillag C, Bjerrum JT, Troelsen JT, et al. Diagnosis of ulcerative colitis before onset of inflammation by multivariate modeling of genome-wide gene expression data. Inflammatory Bowel Diseases. 2009;15(7):1032-8.

9. Wu F, Dassopoulos T, Cope L, Maitra A, Brant SR, Harris ML, et al. Genome-wide gene expression differences in Crohn’s disease and ulcerative colitis from endoscopic pinch biopsies: insights into distinctive pathogenesis. Inflammatory Bowel Diseases. 2007;13(7):807-21.

10. Planell N, Lozano JJ, Mora-Buch R, Masamunt MC, Jimeno M, Ordas I, et al. Transcriptional analysis of the intestinal mucosa of patients with ulcerative colitis in remission reveals lasting epithelial cell alterations. Gut. 2013;62(7):967-76.

11. Arijs I, Li K, Toedter G, Quintens R, Van Lommel L, Van Steen K, et al. Mucosal gene signatures to predict response to infliximab in patients with ulcerative colitis. Gut. 2009;58(12):1612-9.

12. Vanhove W, Peeters PM, Staelens D, Schraenen A, Van der Goten J, Cleynen I, et al. Strong Upregulation of AIM2 and IFI16 Inflammasomes in the Mucosa of Patients with Active Inflammatory Bowel Disease. Inflammatory Bowel Diseases. 2015;21(11):2673-82.

13. Gyorffy B, Molnar B, Lage H, Szallasi Z, Eklund AC. Evaluation of microarray preprocessing algorithms based on concordance with RT-PCR in clinical samples. PloS one. 2009;4(5):e5645.

14. Galamb O, Gyorffy B, Sipos F, Spisak S, Nemeth AM, Miheller P, et al. Inflammation, adenoma and cancer: objective classification of colon biopsy specimens with gene expression signature. Dis Markers. 2008;25(1):1-16.

15. Arijs I, De Hertogh G, Lemmens B, Van Lommel L, de Bruyn M, Vanhove W, et al. Effect of vedolizumab (antialpha4beta7- integrin) therapy on histological healing and mucosal gene expression in patients with UC. Gut. 2018;67(1):43-52.

16. Galamb O, Wichmann B, Sipos F, Spisak S, Krenacs T, Toth K, et al. Dysplasia-carcinoma transition specific transcripts in colonic biopsy samples. PloS One. 2012;7(11):e48547.

17. Penrose HM, Iftikhar R, Collins ME, Toraih E, Ruiz E, Ungerleider N, et al. Ulcerative colitis immune cell landscapes and differentially expressed gene signatures determine novel regulators and predict clinical response to biologic therapy. Sci Rep. 2021;11(1):9010.

18. Newman AM, Liu CL, Green MR, Gentles AJ, Feng W, Xu Y, et al. Robust enumeration of cell subsets from tissue expression profiles. Nat Methods. 2015;12(5):453-7.

19. Brazil JC, Louis NA, Parkos CA. The role of polymorphonuclear leukocyte trafficking in the perpetuation of inflammation during inflammatory bowel disease. Inflammatory Bowel Diseases. 2013;19(7):1556- 65.

20. Catalan-Serra I, Sandvik AK, Bruland T, Andreu- Ballester JC. Gammadelta T Cells in Crohn’s Disease: A New Player in the Disease Pathogenesis? J Crohns Colitis. 2017;11(9):1135-45.

21. Tindemans I, Joosse ME, Samsom JN. Dissecting the Heterogeneity in T-Cell Mediated Inflammation in IBD. Cells. 2020;9(1).

22. Leiper K, Martin K, Ellis A, Subramanian S, Watson AJ, Christmas SE, et al. Randomised placebo-controlled trial of rituximab (anti-CD20) in active ulcerative colitis. Gut. 2011;60(11):1520-6.

23. Goetz M, Atreya R, Ghalibafian M, Galle PR, Neurath MF. Exacerbation of ulcerative colitis after rituximab salvage therapy. Inflammatory Bowel Diseases. 2007;13(11):1365-8.

24. Hart AL, Al-Hassi HO, Rigby RJ, Bell SJ, Emmanuel AV, Knight SC, et al. Characteristics of intestinal dendritic cells in inflammatory bowel diseases. Gastroenterology. 2005;129(1):50-65.

25. Mantovani A, Sica A, Sozzani S, Allavena P, Vecchi A, Locati M. The chemokine system in diverse forms of macrophage activation and polarization. Trends Immunol. 2004;25(12):677-86.

26. Hume DA. The Many Alternative Faces of Macrophage Activation. Front Immunol. 2015;6:370.

27. Rescigno M, Di Sabatino A. Dendritic cells in intestinal homeostasis and disease. J Clin Invest. 2009;119(9):2441-50.

28. Mayer L, Shlien R. Evidence for function of Ia molecules on gut epithelial cells in man. J Exp Med. 1987;166(5):1471-83.

29. Duchmann R, Kaiser I, Hermann E, Mayet W, Ewe K, Meyer zum Buschenfelde KH. Tolerance exists towards resident intestinal flora but is broken in active inflammatory bowel disease (IBD). Clinical and Experimental Immunology. 1995;102(3):448-55.

30. Loktionov A. Eosinophils in the gastrointestinal tract and their role in the pathogenesis of major colorectal disorders. World J Gastroenterol. 2019;25(27):3503-26.

31. Pascal RR, Gramlich TL, Parker KM, Gansler TS. Geographic variations in eosinophil concentration in normal colonic mucosa. Mod Pathol. 1997;10(4):363-5.

32. Oussalah A, Evesque L, Laharie D, Roblin X, Boschetti G, Nancey S, et al. A multicenter experience with infliximab for ulcerative colitis: outcomes and predictors of response, optimization, colectomy, and hospitalization. The American Journal of Gastroenterology. 2010;105(12):2617-25.

33. Sandborn WJ, Rutgeerts P, Feagan BG, Reinisch W, Olson A, Johanns J, et al. Colectomy rate comparison after treatment of ulcerative colitis with placebo or infliximab. Gastroenterology. 2009;137(4):1250-60; quiz 520.

34. Ferrante M, Vermeire S, Katsanos KH, Noman M, Van Assche G, Schnitzler F, et al. Predictors of early response to infliximab in patients with ulcerative colitis. Inflammatory Bowel Diseases. 2007;13(2):123-8.

35. Jakobovits SL, Jewell DP, Travis SP. Infliximab for the treatment of ulcerative colitis: outcomes in Oxford from 2000 to 2006. Alimentary pharmacology & Therapeutics. 2007;25(9):1055-60.

36. Smillie CS, Biton M, Ordovas-Montanes J, Sullivan KM, Burgin G, Graham DB, et al. Intra- and Inter-cellular Rewiring of the Human Colon during Ulcerative Colitis. Cell. 2019;178(3):714-30 e22.

37. Banks C, Bateman A, Payne R, Johnson P, Sheron N. Chemokine expression in IBD. Mucosal chemokine expression is unselectively increased in both ulcerative colitis and Crohn’s disease. J Pathol. 2003;199(1):28-35.

38. Manolakis AC, Kapsoritakis AN, Tiaka EK, Potamianos SP. Calprotectin, calgranulin C, and other members of the s100 protein family in inflammatory bowel disease. Digestive diseases and sciences. 2011;56(6):1601-11.

39. Silosi I, Boldeanu MV, Mogoanta SS, Ghilusi M, Cojocaru M, Biciusca V, et al. Matrix metalloproteinases (MMP-3 and MMP-9) implication in the pathogenesis of inflammatory bowel disease (IBD). Rom J Morphol Embryol. 2014;55(4):1317-24.

40. Taylor CT, Colgan SP. Hypoxia and gastrointestinal disease. Journal of Molecular Medicine. 2007;85(12):1295-300.

41. Moessinger C, Klizaite K, Steinhagen A, Philippou- Massier J, Shevchenko A, Hoch M, et al. Two different pathways of phosphatidylcholine synthesis, the Kennedy Pathway and the Lands Cycle, differentially regulate cellular triacylglycerol storage. BMC Cell Biol. 2014;15:43.

42. McCoy MG, Sun GS, Marchadier D, Maugeais C, Glick JM, Rader DJ. Characterization of the lipolytic activity of endothelial lipase. J Lipid Res. 2002;43(6):921-9.

43. Ahmed K, Tunaru S, Langhans CD, Hanson J, Michalski CW, Kolker S, et al. Deorphanization of GPR109B as a receptor for the beta-oxidation intermediate 3-OHoctanoic acid and its role in the regulation of lipolysis. J Biol Chem. 2009;284(33):21928-33.

44. Peters A, Krumbholz P, Jager E, Heintz-Buschart A, Cakir MV, Rothemund S, et al. Metabolites of lactic acid bacteria present in fermented foods are highly potent agonists of human hydroxycarboxylic acid receptor 3. PLoS Genet. 2019;15(5):e1008145.

45. Bosch M, Sanchez-Alvarez M, Fajardo A, Kapetanovic R, Steiner B, Dutra F, et al. Mammalian lipid droplets are innate immune hubs integrating cell metabolism and host defense. Science. 2020;370(6514).

46. Heller S, Cable C, Penrose H, Makboul R, Biswas D, Cabe M, et al. Intestinal inflammation requires FOXO3 and Prostaglandin E2 dependent lipogenesis and elevated lipid droplets. Am J Physiol Gastrointest Liver Physiol. 2016:ajpgi 00407 2015.

47. Cheng L, Han X, Shi Y. A regulatory role of LPCAT1 in the synthesis of inflammatory lipids, PAF and LPC, in the retina of diabetic mice. Am J Physiol Endocrinol Metab. 2009;297(6):E1276-82.

48. Wang Q, Zhou Y, Rychahou P, Fan TW, Lane AN, Weiss HL, et al. Ketogenesis contributes to intestinal cell differentiation. Cell Death Differ. 2017;24(3):458-68.

49. Cheng CW, Biton M, Haber AL, Gunduz N, Eng G, Gaynor LT, et al. Ketone Body Signaling Mediates Intestinal Stem Cell Homeostasis and Adaptation to Diet. Cell. 2019;178(5):1115-31 e15.

50. Huang LH, Melton EM, Li H, Sohn P, Jung D, Tsai CY, et al. Myeloid-specific Acat1 ablation attenuates inflammatory responses in macrophages, improves insulin sensitivity, and suppresses diet-induced obesity. Am J Physiol Endocrinol Metab. 2018;315(3):E340-E56.

51. Sifroni KG, Damiani CR, Stoffel C, Cardoso MR, Ferreira GK, Jeremias IC, et al. Mitochondrial respiratory chain in the colonic mucosal of patients with ulcerative colitis. Mol Cell Biochem. 2010;342(1-2):111-5.

52. Santhanam S, Rajamanickam S, Motamarry A, Ramakrishna BS, Amirtharaj JG, Ramachandran A, et al. Mitochondrial electron transport chain complex dysfunction in the colonic mucosa in ulcerative colitis. Inflammatory Bowel Diseases. 2012;18(11):2158-68.

53. Yu X, Wieczorek S, Franke A, Yin H, Pierer M, Sina C, et al. Association of UCP2 -866 G/A polymorphism with chronic inflammatory diseases. Genes Immun. 2009;10(6):601-5.

54. Heller S, Penrose HM, Cable C, Biswas D, Nakhoul H, Baddoo M, et al. Reduced mitochondrial activity in colonocytes facilitates AMPKalpha2-dependent inflammation. FASEB J. 2017.

55. Ruiz E, Penrose HM, Heller S, Nakhoul H, Baddoo M, Flemington EF, et al. Bacterial TLR4 and NOD2 signaling linked to reduced mitochondrial energy function in active inflammatory bowel disease. Gut microbes. 2019:1-14.

56. Mancini NL, Goudie L, Xu W, Sabouny R, Rajeev S, Wang A, et al. Perturbed Mitochondrial Dynamics Is a Novel Feature of Colitis That Can Be Targeted to Lessen Disease. Cell Mol Gastroenterol Hepatol. 2020;10(2):287- 307.

57. Lee JY, Cevallos SA, Byndloss MX, Tiffany CR, Olsan EE, Butler BP, et al. High-Fat Diet and Antibiotics Cooperatively Impair Mitochondrial Bioenergetics to Trigger Dysbiosis that Exacerbates Pre-inflammatory Bowel Disease. 2020;28(2):273-84 e6.

58. Hession C, Osborn L, Goff D, Chi-Rosso G, Vassallo C, Pasek M, et al. Endothelial leukocyte adhesion molecule 1: direct expression cloning and functional interactions. Proc Natl Acad Sci U S A. 1990;87(5):1673-7.

59. Rajca S, Grondin V, Louis E, Vernier-Massouille G, Grimaud JC, Bouhnik Y, et al. Alterations in the intestinal microbiome (dysbiosis) as a predictor of relapse after infliximab withdrawal in Crohn’s disease. Inflammatory bowel diseases. 2014;20(6):978-86.

Author Information X