Summary
The incidence of inflammatory bowel diseases (IBD) worldwide has resulted in a global public health challenge. Intestinal fibrosis leading to stricture formation and bowel obstruction is a frequent complication in Crohn’s disease (CD), and the lack of anti-fibrotic therapies makes elucidation of fibrosis mechanisms a priority. Progress has shown that mesenchymal cells, cytokines, microbial products and mesenteric adipocytes are jointly implicated in the pathogenesis of intestinal fibrosis. This recent information puts prevention or reversal of intestinal strictures within reach through innovative therapies validated by reliable clinical trial endpoints. Here, we review the role of immune and non-immune components of the pathogenesis of intestinal fibrosis, including new cell clusters, cytokine networks, host-microbiome interactions, creeping fat, and their translation for endpoint development in anti-fibrotic clinical trials.
Keywords: Intestinal fibrosis, inflammatory bowel disease, myofibroblasts, single cell RNA (scRNA) sequencing, cytokine, creeping fat
Introduction
Inflammatory bowel disease (IBD), including ulcerative colitis (UC) and Crohn’s disease (CD), is a relapsing inflammatory condition of the gastrointestinal tract. It has become a healthcare burden with the highest prevalence in Europe and North America, and accelerating incidence in Asia, Africa and South America 1. While UC is restricted to the colon with continuous superficial inflammation of the mucosa layer, CD can occur in both small and large intestine with patchy, transmural inflammation 2. Intestinal fibrosis which results in obstruction and need for surgical interventions is a serious and common complication of CD 3. Deposition of extracellular matrix (ECM) is part of physiologic tissue repair and restitution after injury. However, in states of chronic inflammation such as CD, that are associated with persistent inflammation lasting for months or years, progressive fibrosis with excessive accumulation of ECM, hardening and scarring of intestinal tissues ensues, leading to impaired function and organ damage 4. Fibroblasts and myofibroblasts are the primary ECM producers, but emerging evidence points towards additional mechanisms of gut obstruction. In fact, thickening of the intestinal muscularis propria appears to be the major contributor to luminal narrowing and development of clinical symptoms 5. In their lifetime more than half of CD patients develop clinically apparent strictures, which are characterized on cross sectional imaging or histopathology by the presence of intestinal luminal narrowing, induced by bowel wall thickening 6. However, in these population-based studies, diagnosis of strictures is mostly based on symptoms, and likely underestimates the real incidence of fibrosis.
Despite awareness of its clinical importance in CD, fibrogenesis and stricture formation, have been largely overlooked in UC. Colonic fibrosis in UC has significant clinical implications, including motility abnormalities, anorectal dysfunction, rectal urgency, and incontinence 7. Stricture formation is an infrequent event in UC [1–10%] as compared with small bowel CD, but interestingly, rates of colonic strictures are comparable in CD and UC. UC is generally acknowledged to be an inflammatory process confined to the mucosal/submucosal layers. However, in a study with 706 tissue cross-sections derived along the length of 89 consecutive UC colectomy specimens and compared to CD, diverticular disease and normal areas from colorectal cancer specimens, submucosal fibrosis was detected in 100% of UC specimens 8. Submucosal fibrosis and thickening of the muscularis mucosae were associated with severe and chronic injury, but not active inflammation, suggesting fibrosis and muscularis mucosae thickening are common complications of chronic progressive UC 8. Another recent investigation showed persistent abnormalities even in endoscopically healed UC mucosa, including ECM remodeling, profibrotic cytokines production, and activated TGF-β signaling pathways, again suggesting that while inflammation is necessary to initiate fibrogenesis, suppression of inflammation is not sufficient to prevent intestinal fibrosis 9. The fact that most experimental models of intestinal fibrosis have a colonic location 10 and most preclinical target validation occurs in these models, supports the notion that intestinal fibrosis occurs in areas of inflammation irrespective of disease location.
In this review, we highlight cutting-edge advances in the field of intestinal fibrosis, including single cell RNA (scRNA) sequencing, novel cytokines, and aspects unique to the intestine that have therapeutic potential, namely the microbiota and the interaction of intestinal muscle with mesenteric fat. We also highlight a pathway for translation of these findings into clinical practice.
Overview of IBD pathogenesis
Although the origin of IBD remains incompletely understood, several studies have demonstrated that complex interactions between genetic predisposition and environmental factors have a crucial role in the development of this chronic disease 11. Genome-wide association studies have identified more than 200 loci associated with the risk of CD or UC, including NOD2, ATG16L1, CARD9 and IL23R 12–14. It is worth noticing that many of these IBD risk loci are found in regions encoding molecules that regulate cytokine signaling, immune response to microbes and autophagy signaling 15–17, indicating the essential role of cytokine and host-microbe interactions in IBD pathogenesis. Environmental risk factors (collectively grouped under the rubric “exposome”), including smoking, diet, food additives, the use of antibiotics, air and water pollution are likely also involved in the onset or progression of IBD 18. These exposome factors interact and likely alter the composition of the gut microbiome, which plays a major role in human health and is a key factor in IBD pathogenesis 19. The interplay between genetic and environmental factors leads to impaired epithelial barrier function, resulting in the translocation of microbial products into the bowel wall, where they first are recognized by innate immune cells such as dendritic cells and macrophages 20. Once activated these cells produce cytokines and chemokines, resulting in recruitment of additional immune cells and initiation of adaptive immune responses 21,22. The excessive and uncontrolled innate and adaptive responses trigger chronic intestinal inflammation, further impairment of barrier function, gut tissue damage, persistence of IBD, and fibrogenic events 23.
Mechanisms and cellular components of intestinal fibrogenesis
Fibrosis develops to preserve tissue architecture and functional integrity, and is an essential component of wound healing and tissue repair that usually follows tissue injury and associated inflammation 24,25. However, prolonged exposure to injurious stimuli may lead to progressive fibrosis characterized by excess deposition of ECM, scarring of various tissues, impaired function and organ damage 26,27. A majority of IBD patients, particularly those with CD, will develop intestinal fibrosis, a serious and common complication that ultimately results in obstruction and surgical interventions 28. Intestinal mesenchymal cells, including fibroblasts, myofibroblasts and smooth muscle cells are the main cell types responsible for ECM secretion, and the combined increase of mesenchymal cell numbers and excessive ECM secretion are considered the hallmark features of intestinal strictures 4. Other than secretion, ECM accumulation is also regulated by matrix metalloproteinases (MMPs) and their inhibitors, the tissue inhibitors of metalloproteinases (TIMPs). MMPs are proteolytic enzymes that break down ECM, while TIMPs counteract their degrading activity. An imbalance between MMPs and TIMPs may lead to ECM accumulation and subsequently fibrosis 29. Here we will discuss the main cell types and cell differentiation mechanisms involved in ECM secretion and fibrosis in the intestine (Figure 1).
Mesenchymal cells.
Mesenchymal cells are non-epithelial, non-endothelial, non-haematopoietic cells that differentiate from mesenchymal stem cells and act as a scaffold for tissue structure although they also exert other functions 30. In the intestine, mesenchymal cells are found in all anatomical compartments and most of them participate in the process of intestinal fibrosis, including intestinal subepithelial myofibroblasts, lamina propria fibroblasts, pericytes, bone marrow–derived mesenchymal stromal or stem cells, and smooth muscle cells (SMC), as well as the interstitial cells of Cajal and fibrocytes, the latter usually considered non-conventional mesenchymal cells 31.
Fibroblasts, smooth muscle cells and myofibroblasts.
According to the current marker-based classification system, intestinal fibroblasts express the surface marker CD90 and the intermediate filament protein vimentin, but not desmin, whereas smooth muscle cells are CD90 (−), vimentin (low), α-SMA (+), and desmin (+) 32. Myofibroblasts are CD90 (+), vimentin (+), α-SMA (+), and desmin (− or low), granting them an intermediate phenotype between fibroblasts and smooth muscle cells 31. In the inflamed gut, resident mesenchymal cells actively differentiate and dedifferentiate between these three interrelated mesenchymal cell phenotypes 33. In case of acute injury or inflammation, fibroblasts are activated, migrate to and proliferate at the site of injury and contribute to ECM production. Upon stimulation with specific growth factors such as TGF-β1, they start expressing α-SMA and differentiate into myofibroblast-like cells 34,35. Similarly, smooth muscle cells can also transdifferentiate into myofibroblasts 36. However, upon stimulation by pro-inflammatory cytokines, myofibroblasts can differentiate back into smooth muscle cells, induce SMC hyperplasia/hypertrophy, leading to thickened muscularis propria, stricture formation and intestinal obstruction 37. Accumulating evidence suggests that myofibroblasts are the primary producers of ECM 38. Myofibroblasts can be divided into two main types, interstitial cells of Cajal and intestinal subepithelial myofibroblasts. Interstitial cells of Cajal reside in the submucosa and the muscularis propria and provide signals that facilitate gastrointestinal motility, whereas intestinal subepithelial myofibroblasts are located within the lamina propria directly under only one layer of epithelial cells 38. Subepithelial myofibroblast are dense in the crypt region and sparse at the colonic surface and the villi of the small bowel 38. In IBD chronic inflammation fosters production of inflammatory and profibrotic mediators that sustain the activation and proliferation of myofibroblasts, leading to ECM overproduction and fibrosis 39.
Fibrocytes.
Fibrocytes are bone marrow-derived mesenchymal stem cells 40. Under certain circumstances, fibrocytes circulating in the peripheral blood travel to affected tissues where they differentiate into fibroblasts/myofibroblasts upon stimulation by inflammatory and pro-fibrotic growth factors 41. Fibrocytes may enhance inflammation by producing TNF-α and can directly augment fibrosis by producing ColI 42. In CD patients, levels of circulating fibrocytes are increased compared to healthy controls 42,43, but circulating fibrocytes can also be elevated in UC patients 44. To date there is no evaluation of fibrocytes function in intestinal fibrosis.
Pericytes.
Another type of ECM-producing cells are pericytes, which are found surrounding blood vessels, adjacent to endothelial cells 38, and in histological sections they display the shape of myofibroblasts 45. Pericytes are defined as α-SMA(−) and desmin-positive cells, exhibiting an intermediate phenotype between vascular smooth muscle cells and fibroblasts 31. Under acute or chronic inflammatory conditions, pericytes differentiate into fibroblasts by losing their original markers, expressing typical fibroblast markers and, by producing large quantities of ECM components 46, may contribute to intestinal fibrosis. Increased numbers of activated pericytes are found in the inflamed mucosa of patients with long lasting UC 47. However, the exact role of pericytes in intestinal fibrosis remains unclear.
Epithelial-Mesenchymal Transition (EMT).
EMT represents a cellular trans-differentiation event where typical epithelial cell markers like adherens and tight junctions (E-cadherin, ZOs and claudins) and cytoskeletal proteins (cytokeratins) are lost in epithelial cells, which then acquire mesenchymal neural cadherin (N-cadherin) expression together with the upregulation of α-SMA, vimentin, fibronectin, collagens and the transcription factors Twist, Snail, Slug, Zeb1/2. Along with these phenotypic changes, these events help transitioning stationary epithelial cells to migratory and invasive cells 48,49. EMT was first described in organs such as the kidney, liver and lung, and contribute to the fibrogenic process by generating epithelial-derived fibroblasts that become responsible for ECM deposition and tissue scarring 50. Later, a role for EMT in the pathogenesis of IBD-associated intestinal fibrosis also emerged 50. A recent study demonstrated the occurrence of EMT in fibrotic intestinal CD tissue independent of the effect of inflammation 51. Another report detected fibrosis and expression of EMT markers in samples from CD patients, irrespective of the clinical behavior [stricturing (Montreal classification: B2) or penetrating (Montreal classification: B3) behavior] 52. The functional role of EMT in IBD still needs to be determined.
Endothelial-Mesenchymal Transition (EndoMT).
EndoMT can be viewed as EMT occurring in endothelial cells. Similarly to epithelial cells, during the process of EndoMT endothelial cells lose the expression of typical endothelial markers and acquire fibroblast-like morphology and gene expression profile 53. Primary human intestinal microvascular endothelial cells (HIMECs) undergo EndoMT when exposed to TGFβ, IL-1β and TNFα, or supernatants from lamina propria primary mononuclear cells 54. During the process of EndoMT, HIMECs acquire an elongated morphology and broad changes in the gene expression profile, including downregulation of genes typically expressed by endothelial cells (CD31, VE-cadherin, vWF), downregulation of ECM components normally produced by endothelial cells (Collagen IV, entactin) and, conversely, upregulation of genes encoding ECM components expressed during intestinal fibrosis (Collagen I, fibronectin) 54. Evidence of in vivo EndoMT can be found in human colonic tissues from both CD and UC patients 54. EndoMT is also involved in radiation-induced rectal fibrosis, and in mucosal and submucosal vessels is proportional to the radiation injury score of patients with radiation-induced proctitis 55.
Activated fibroblast clusters and intestinal fibrosis
While traditional mesenchymal cell phenotypes have strongly contributed to the field of intestinal fibrosis, recent single cell genomics approaches are transforming our knowledge on fibrotic disease across various organ systems 56. Its unprecedented resolution allows the interrogation of cell types and functional states with unmatched accuracy, opening a new window into disease mechanisms. Little is known about the application of single cell sequencing (scRNA-seq) in intestinal fibrosis. However, scRNA-seq analysis from other organs, such as lung, liver, skin, kidney and heart, provide new clues about cell clusters related to fibrosis and will help to guide the establishment of scRNA-seq in intestinal fibrosis studies.
In the lung, using single-cell transcriptome analyses, a new mesenchymal cells subtype expressing high levels of Pdgfrb was identified, with increased Pdgfrb expression in fibrotic mesenchymal cells compared to normal mesenchymal cells 57. scRNA-seq of alveolar fibroblasts also revealed a subset expressing Wnt5a scattered throughout the alveolar region, most near an Axin2+ AT2 cell 58. In liver fibrosis, hepatic stellate cells (HSC) are considered the major source of myofibroblasts 59,60, with several scRNA-seq studies describing the HSC/myofibroblasts and immune cell populations involved. In a study using isolated HSCs and activated myofibroblasts from the carbon tetrachloride (CCl4)-induced liver fibrosis mouse model, S100 calcium binding protein A6 (S100A6) was found to be a universal marker of activated myofibroblasts, but heterogeneity was noticed in both HSCs and activated myofibroblasts, indicating the existence of functionally relevant subsets in hepatic fibrosis 61. Pathogenic subpopulations of TREM2+CD9+ macrophages, ACKR1+ and PLVAP+ endothelial cells and PDGFRα+ collagen-producing myofibroblasts were also identified, revealing intra-scar activity of several pro-fibrogenic pathways, including TNFRSF12A, PDGFR and NOTCH signaling 62. Central vein-associated HSCs were described as the dominant collagen-producing cells in a mouse model of centrilobular fibrosis, and LPAR1 was identified as a therapeutic target for collagen-production in a rodent liver fibrosis non-alcoholic steatohepatitis NASH model 63. scRNA-seq also revealed the contribution of monocytes to myofibroblasts in kidney fibrosis 64. Two distinct populations of myofibroblasts were identified, resident myofibroblasts (PDGFRβ+CD45−) largely associated with ECM production, and circulating cells-derived myofibroblasts (PDGFRβ+CD45+) which exhibit a monocyte-like phenotype associated with immune responses 64. Another study used both single-cell RNA sequencing and single-nucleus RNA sequencing to uncover novel cell types and cell states related to kidney fibrosis 65. In this study, two distinct α-SMA positive activated fibroblast populations were identified: mannose receptor 2-expressing renal myofibroblasts, which bind and internalize collagen and attenuate renal fibrosis, and tenascin C (an extracellular glycoprotein)-expressing renal myofibroblasts 65 that promote renal fibrosis 66.
scRNA-seq technologies have also been applied to IBD. A recent study of the colonic mesenchyme revealed four distinct subsets of fibroblasts in addition to pericytes and myofibroblasts 67. The mesenchymal niches are dysregulated in intestinal inflammation with appearance of an activated fibroblast population expressing interleukin (IL)-33, lysyl oxidases (LOX), TNFSF14 and fibroblastic reticular cell-associated genes, leading to impaired epithelial function. This highlights novel types of fibroblasts and how intestinal fibroblasts drive inflammation and barrier dysfunction in IBD 67. In the context of ileal CD cellular heterogeneity may contribute to resistance to treatment with anti-TNF therapy. 82,417 lamina propria cells from 22 samples were sequenced in one study and interestingly the tissue in the anti-TNF resistant group contained two subsets of fibroblasts, one of which was characterized by an activation program including strong expression of THY1 (CD90), PDPN (podoplanin), CTHRC1 (collagen triple-helix repeat-containing 1), and CHI3L1 68. CTHRC1+ fibroblasts are of particular interest, since this cell population expresses the highest amount of collagen, can be found in fibroblast foci in idiopathic pulmonary fibrosis (IPF), and show high migration and invasion capacity in vitro 69. However, the role of these activated fibroblasts or other mesenchymal cell populations in the pathogenesis of intestinal fibrosis is not yet clear. Discovery of spatially discrete, functionally unique fibroblast subsets with non-overlapping functions will have a critical impact in the rational design of therapies aimed at precisely modulating inflammation, fibrosis and tissue repair.
Cytokine networks and immune cells in intestinal fibrosis
In IBD mucosal fibroblasts and myofibroblasts in the mucosa are exposed to a highly complex microenvironment consisting of multiple cytokines released by several different cell types, with immune cells in the lamina propria being the most important sources. Investigation in both humans and animal models suggests that the release of cytokines during chronic inflammation could trigger the onset and directly promote intestinal fibrosis 70. However, it remains unclear whether the neutralization of a single cytokine or a combination of several cytokines will prevent or even reverse intestinal fibrosis. Hence, it is worth discussing the cytokines related to intestinal fibrosis and the main cell types releasing these cytokines, particularly newly described pro-fibrotic factors such as IL-11, IL-33, IL-34 and IL-36 (Figure 2).
TGF-β.
TGF-β is considered the key driver of intestinal fibrosis in IBD. In 1990 it was reported that TGF-β1 could selectively augment collagen production by human intestinal smooth muscle cells in vitro 71. Following studies elucidated the overexpression of TGF-βs and their signaling receptors in tissue samples from intestinal CD resections, indicating a potential role of these regulatory molecules in the pathophysiology of CD 72. Furthermore, the expression levels of TGF-β transcripts and phosphorylated Smad2/3 were elevated in the mucosa overlying strictures but not non-strictured areas in CD tissues 73. Consistently, myofibroblasts from mucosa overlying strictured gut also showed higher expression of TGF-β transcripts than myofibroblasts from mucosa overlying non-strictured gut 73.
After binding to its receptor, TGF-β activates intracellular signaling through both the Smad-dependent and Smad-independent pathways. In the Smad-dependent pathway, the TGF-β receptor complex directly phosphorylates Smad2 and Smad3, which then forms a complex with Smad4 and enters the nucleus to regulate the transcription of target genes 74. Smad6 and Smad7, which compete with Smad2 and Smad3 for TGF-β receptor I kinase and promote the degradation of the TGF-β receptor, negatively regulate the TGF-β/Smad2–3 signaling pathway 75. Additionally, Smad7 was reported to have a lower expression level in the mucosa overlying strictured than non-strictured areas 73. Smad-independent TGF-β signaling pathways, including mitogen-activated protein kinase (MAPK) and the phosphatidylinositol-3-kinase (PI3K) cascade, can also be activated by TGF-β directly 76. Activated TGF-β promotes the differentiation from fibroblasts to myofibroblasts, resulting in elevated expression of α-SMA, increased proliferation and decreased apoptosis 38. In addition, TGF-β treatment enhances the migration and collagen production of myofibroblasts 77,78. In human intestinal organoids which contain both epithelial and mesenchymal cells, including myofibroblasts, TGF-β stimulation increases the production of a series of pro-fibrotic factors, such as collagen I, α-SMA and fibronectin 79. In animal models, TGF-β1 peptide-based vaccine suppresses excessive TGF-β1 bioactivity and ameliorates fibrosis in a mouse model of chronic colitis 80. Knockdown of Smad7 with a specific antisense oligonucleotide attenuated colitis and colitis-driven colonic fibrosis in a 2, 4, 6-trinitrobenzenesulfonic acid (TNBS) mice model 81. These results may result from the many different effects of TGF-β in inflammation and fibrosis, which poses a challenge in generating anti-fibrotic treatments targeting TGF-β. Recently, several new proteins and factors have been reported to have a role in intestinal fibrosis through regulation of the TGF-β signaling pathway, such as ER stress protein glucose-regulated protein (GRP)78 82, fibrin 83, Cadherin-11 84, anexelekto (AXL) 85, nuclear factor erythroid 2–related factor 2 (Nrf2) 86 and others.
IL-1 family members.
There are 11 members of the IL-1 family of cytokines, which can be divided into 3 subfamilies: the IL-1 subfamily (containing IL-1α, IL-1β, IL-33, and IL-1Ra), IL-18 subfamily (containing IL-18 and IL-37), and IL-36 subfamily (containing IL-36Ra, IL-36α, β, γ and IL-38) 87. Besides the well-established inflammatory properties of IL-1 family members, some of them, such as IL-1, IL-33 and IL-36, also have an emerging role in intestinal fibrosis.
IL-1α and IL-1β
IL-1α can function via its cell surface receptor IL-1R1 or regulate gene expression directly 88. IL-1α is constitutively expressed in epithelial cells, but also can be expressed by other cell types upon their activation, such as macrophages, monocytes, endothelial cells and others 88. IL-1α released from damaged intestinal epithelial cells acts as a damage-associated molecular pattern (DAMP), contributing to the initiation and development of chronic intestinal inflammation 38. In a DSS-induced colitis mouse model, IL-1α-deficient mice exhibited mild disease symptoms with improved recovery, and neutralization of IL-1α in control mice during acute colitis led to alleviation of clinical and histological manifestations. IL-1β deficiency correlated with disease exacerbation and treatment with rIL-1Ra or anti-IL-1β antibodies was not effective in DSS-induced acute colitis mice model 89. Intestinal epithelial cell lysate-induced activation of human intestinal fibroblasts (HIFs) is mediated by IL-1R and IL-1α but not IL-1β 90. Furthermore, intestinal epithelial cell-derived IL-1α induces cytokine production by HIFs 90. IL-1α and TNF-α also increased TGF-β1 and TIMP-1 production by colonic epithelial cells 77. In other organs, IL-1α also acts as a profibrotic cytokine, as IL-1α-deficient mice exhibit reduced collagen deposition in response to bleomycin treatment in lung fibroblasts 91.
IL-1β is mainly produced by mononuclear phagocytes, acting as a pro-inflammatory effector in intestinal inflammation 70. IL-1β enhances TGF-β1-induced EMT in human bronchial epithelial cells 92. IL-1β over-expression induces acute lung injury and chronic repair leading to pulmonary fibrosis in a rat model and the IL-1R1/MyD88 axis is considered essential in pulmonary inflammation and fibrosis in mice 93–95. In liver fibrosis, although IL-1β promoted HSC proliferation and was associated with the degree of liver fibrosis in Abcb4−/− mice, IL-1 antagonism showed anti-fibrotic effects in vitro but not in Abcb4−/− mice 96. IL-1β induces a metabolic switch in platelet-derived growth factor receptor (PDGFRβ) + kidney stromal cells, which promotes tubulointerstitial fibrosis 97. IL-1β stimulates the secretion of collagens I and IV, IL-8, monocyte chemoattractant protein (MCP)-1 and MMP-1 by colonic subepithelial myofibroblasts 98. However, another report suggested IL-1β inhibited collagen synthesis and induced collagenase and TIMP-1 production in intestinal smooth muscle cells 99,100. The role of IL-1β in intestinal fibrosis remains unclear.
IL-36
IL-36 consists of three agonists, IL-36α (IL-1F6), IL-36β (IL-1F8), IL-36γ (IL-1F9), and IL-36 receptor antagonist (IL-36Ra) 101,102. The IL-36 agonists have demonstrated pro-inflammatory effects and recent evidence suggests additional roles in intestinal fibrosis 101,102. In the intestinal tract, IL-36 is generated by many different cell types, including fibroblasts, myofibroblasts, epithelial cells, goblet cells, macrophages, and glial cells 103–108. In turn, many cell types can respond to IL-36, including immune cells, epithelial cell, endothelial cells, and mesenchymal cells, and IL-36 induces distinct gene expression and functional changes 109. IL‐36α and IL-36γ were upregulated in endoscopic biopsies of patients with active colonic CD compared to unaffected biopsies from the same patients 110. In addition, increased expression of IL-36α and IL-36γ, but not of IL-36β, was observed in the intestinal mucosa in UC patients rather than control or CD patients 107,108. A pro-fibrotic role of IL-36 has been suggested in pulmonary fibrosis and renal fibrosis 88,111. In the intestine, increased IL36A expression was also found in tissues from patients with fibrostenotic CD, with high numbers of activated myofibroblasts 112. In both mouse and human fibroblasts, IL36R activation enhances the expression of genes that regulated fibrosis and tissue remodeling, with higher expression of collagen VI 112. IL-36R-deficient mice or mice injected with anti-IL-36R antibody develop less severe colitis and fibrosis in DSS- or TNBS-induced colitis 112, suggesting that blockade of IL36R signaling has the potential to be an efficient mechanism for prevention and treatment of intestinal fibrosis in IBD patients.
IL-33
IL-33 is expressed throughout various organs in the body and can be produced by both non-hematopoietic and hematopoietic cells, including fibroblasts, adipocytes, smooth muscle cells, endothelial cells, bronchial, intestinal epithelial cells, macrophages and dendritic cells 113. IL-33 signals through its receptor ST2 and drives the production of pro-inflammatory and type 2 helper T cell (Th2)-associated immune responses, which lead to the development of fibrotic diseases in various organs, such as the lung, liver, kidney, heart, skin, and pancreas in an ST2- and macrophage-dependent manner, regulating EMT, or contributing to abnormal fibroblast proliferation, leukocyte infiltration and morphologic differentiation of human endothelial cells 114. In the gut, IL-33 is mainly expressed by fibroblasts, smooth muscle cells, endothelial cells, and adipocytes, as well as intestinal epithelial cells and infiltrating lamina propria mononuclear cells in active UC 113. Increased expression levels of IL-33 and its receptor, ST2, was found in IBD patients, particularly in UC 115,116. In adherent invasive E-coli (AIEC)-elicited experimental intestinal fibrosis, the bacterial protein flagellin potentiated the expression of the IL-33 receptor ST2 in the intestinal epithelium 117. The IL-33-ST2 axis promoted intestinal fibrosis in a TLR5-and NOD-like receptor family CARD domain-containing protein (NLRC) 4-dependent manner. Blockade of IL-33-ST2 signaling by an anti-ST2 blocking antibody suppresses fibrosis-related gene expression and the accumulation of collagen in the colon 117. Myd88, which is known for its essential role in TLR signaling pathways as an adaptor protein in innate immune responses, was demonstrated to act downstream TLR5 and regulate intestinal fibrosis 118. In a α-SMA-specific MyD88 deletion mouse model, MyD88 deletion prior to, but not after induction of, experimental colitis resulted in decreased intestinal fibrosis 118. α-SMA(+) HIMFs selectively respond to flagellin with enhanced fibronectin or Col1 production in an MyD88-dependent manner, mediated by eIF2 alpha and 4EBP1 118. These results further indicate signaling pathways involved in flagellin induced intestinal fibrosis.
IL-6 family members.
The interleukin (IL)-6 family of cytokines includes IL-6, IL-11, ciliary neurotrophic factor (CNTF), leukemia inhibitory factor (LIF), oncostatin M (OSM), cardiotrophin 1 (CT-1), cardiotrophin-like cytokine (CLC), and IL-27 119, all of which utilize a common signaling receptor subunit glycoprotein 130 kDa (gp130). Blockade of IL-6 family cytokines has been shown to be beneficial in autoimmune diseases, but bacterial infections and metabolic side effects have been observed 119. Current studies suggest IL-6 and IL-11 may have a role in intestinal fibrosis.
IL-6
IL-6 can be expressed by intestinal mononuclear cells, epithelial cells and mesenchymal cells, especially upon challenge with inflammatory cytokines 70, and the role of IL-6 is well established in IBD 120. Besides its known pro-inflammatory activity, IL-6 also contributes to fibrosis. High levels of IL-6 characterize early idiopathic pulmonary fibrosis acute exacerbations and an increase in the levels of IL-6 associates with worse outcome in idiopathic pulmonary fibrosis patients 121. The HLF/IL-6/STAT3 circuit drives HSC activation in liver fibrosis and IL-6 and p-STAT3 levels were increased in patients with fibrotic livers 122. Upon TGF-β activation, intestinal mesenchymal cells are activated and increase their autocrine production of a number cytokines including IL-6, as well as excess ECM production including collagen I 123. In the strictured intestine, immunoreactive IL-6 is increased in α-SMA(+) muscle cells compared with the normal resection margin in structuring (Montreal classification: B2) CD patients. Treatment of normal muscle cells with IL-6 phenocopied muscle cells from strictured intestine whereas neutralization of autocrine IL-6 reversed STAT3 phosphorylation and normalized expression of TGF-β1 in strictured intestinal muscle 124. The pro-fibrotic role of IL-6 in vivo needs further clarification.
IL-11
IL-11 was characterized as a profibrotic cytokine secreted by myofibroblasts and damaged epithelial cells as well as smooth muscle cells 125,126. Its receptor, IL-11RA, is highly expressed on stromal cells, including fibroblasts, smooth muscle cells, adipocytes, and hepatic/pancreatic stellate cells or pericytes, and also on epithelial/polarized cells, such as hepatocytes, alveolar epithelial cells, and kidney tubular epithelial cells 127,128. IL-11 signaling drives myofibroblast activation, parenchymal cell dysfunction, inflammation and promotes fibrosis in several organs, including lung, liver, heart and skin 127–129. In UC, an inflammation-associated fibroblast subset was identified with elevated expression of IL-11, IL-24, and IL-13RA2 130. Transgenic mice (Il11SMC) with smooth muscle cell-specific conditional overexpression of murine IL-11 develop loose stools, progressive bleeding and rectal prolapse, with inflamed, fibrotic and a thickened bowel wall, accompanied by activation of ERK and STAT3 125. A similar phenotype was observed in a second model with fibroblast-specific expression of IL-11, indicating that both mesenchymal cell types are mechanistically important in driving fibro-inflammation and an inflammatory bowel phenotype in mice 125. However, additional studies using a loss-of-function approach for IL-11 are needed, such as using IL-11 deficient mice or IL-11 neutralizing antibodies, to further determine the impact of IL-11 on intestinal fibrosis.
TNF-α family members
Two TNF-α family members, TNF-α and TL1A, have an established role in the pathogenesis of IBD 70,131. Accumulating evidence suggests they may also have direct effects on myofibroblasts or EMT, and thus be related to fibrosis in the intestine and other organs 70,131.
TNF-α
Anti-TNF blocking monoclonal antibodies have a well-established role in IBD therapy 132,133. TNF-α induces an increase in IL-8, MCP-1 and MMP-1, as well as a change in type I and IV collagen secretion in subepithelial myofibroblasts, but does not affect their proliferation 98. TNF-α upregulates TGF-β and TIMP-1 in colonic epithelial cells and conditioned medium from epithelial cells pretreated with TNF-α induces MMP-9 in colonic subepithelial myofibroblasts 77. The anti-TNF antibody Infliximab induces a significant dose-dependent increase in TIMP-1 production in CD myofibroblast with enhanced myofibroblast migration and decreased collagen production 134. In a peptidoglycan–polysaccharide (PG-PS) induced enterocolitis model with granulomatous inflammation that leads to prominent fibrosis mimicking CD, rat-specific anti-TNF-α treatment prevents inflammation and fibrosis 135. In a study evaluating the possible effect of anti-TNF-α antibodies on small bowel stenosis, no progression of pre-existing stenoses, no appearance of new ones, and complete regression of 1/22 (4.5%) small bowel stenoses was observed after the induction phase 136. However, in another study with pediatric CD patients, those who received early anti-TNFα therapy were less likely to have penetrating but not stricturing complications than those who did not receive early anti-TNFα therapy 137. It is generally considered that anti-TNF antibodies do not prevent strictures in CD 131.
TL1A
TL1A, a protein encoded by TNFSF15, binds to death domain receptor 3 (DR3) and is expressed by a variety of cell types including immune cells, epithelial cells, and fibroblasts. TL1A was originally described as a T cell-costimulatory cytokine, but later found to affect multiple cell lineages with cell-type specific effects, such as proliferation, cytokine secretion or cell differentiation among others 138,139. TL1A mRNA and protein expression is upregulated in IBD, particularly in areas involved by CD 140. TL1A may contribute to IBD pathogenesis via local but not systemic induction of IL-17A, but not IL-4, IL-13 or IFN-γ 141. In human translational studies, CD patients with higher peripheral TL1A expression also exhibited intestinal strictures and worsened ileocecal inflammation with relative sparing of rectosigmoid inflammation 142. Constitutive expression of TL1A in either lymphoid or myeloid cells in transgenic mice leads to enhanced intestinal and colonic fibrosis 143,144. In the DSS and adoptive T cell transfer models there is proximal migration of colonic inflammation, worsen patchy intestinal inflammation and long gross intestinal strictures in TL1A transgenic mice compared to wild-type littermates, with increased T-cell activation markers and interleukin-17 expression 142. Furthermore, TL1A-mediated intestinal fibrosis and fibroblast activation are dependent on specific microbial populations 139. Anti-TL1A antibody can reduce intestinal inflammation and fibrosis by inhibiting the activation of intestinal fibroblasts and reducing the collagen synthesis in the T cell transfer model of chronic colitis 145. In established murine colonic fibrosis, treatment with neutralizing TL1A antibodies results in lowered expression of TGF-β1, with reduced number of fibroblasts and myofibroblasts 146. Primary intestinal myofibroblasts express DR3 and functionally respond to direct TL1A signaling by increasing collagen 146. In a recent study, TL1A overexpressing naïve T cells were transferred into Rag−/−, Rag−/− mice lacking DR3 in all cell types (Rag−/−Dr3−/−), or Rag−/− mice lacking DR3 only on fibroblasts (Rag−/−Dr3ΔCol1a2) to induce colitis and fibrosis, and the results showed that Rag−/−Dr3−/− developed decreased inflammation and fibrosis, and despite similar clinical disease and inflammation as Rag−/−, Rag−/−Dr3ΔCol1a2 exhibited reduced intestinal fibrosis and attenuated fibroblast activation and migration 147.
IL-17 and Th17 cells.
It is generally believed that Th1-cell-associated cytokines drive inflammation, whereas uncontrolled type 2 and type 17 cell responses might drive tissue fibrosis through the excessive deposition of ECM 70. This implies a switch in immunophenotype over time, possibly driven by chronic inflammation itself.
It is becoming increasingly clear that T-cell phenotypes are more heterogenous than previously anticipated. There is evidence demonstrating the regulatory role of IL-17 in the development of fibrosis in multiple organs, such as the liver, skin, lung and heart 38,131, but its contribution to IBD and intestinal fibrosis remains controversial 148. IL-17 cytokines, primarily produced by Th17 cells, consists of six related proteins: IL-17A (also called IL-17), IL-17B, IL-17C, IL-17D, IL-17E (also called IL-25), and IL-17F, which signal through five receptor subunits IL-17RA-IL-17RE to149. In humans, IL-6, IL-21, IL-23, IL-1β, and TGF-β are critical for Th17 development and, in addition to IL-17A-F, Th17 cells also produce IL-21, IL-22, IL-9, IL-26, TNF-α, and chemokine (C-C motif) ligand 20 (CCL20) 148. Th17 cell numbers and Th17-related cytokines, including IL-17, IL-21 and IL-22, are significantly increased in active IBD 150. IL-17A is significantly overexpressed in CD strictured areas compared with non-strictured areas 151. In addition, IL-17A significantly inhibits myofibroblast migration and promotes the production of MMP-3, MMP-12, TIMP-1 and collagen by myofibroblasts from strictured CD tissues 151. In subepithelial myofibroblasts, IL-17A enhances the production of collagen I and heat shock protein 47 (HSP47), which is significantly elevated in the intestinal tissues of patients with active CD and contributes to IL-17A-induced collagen I expression 152. Occurrence of EMT and high levels of IL-17A are observed in the intestinal mucosa of CD patients and IL-17A may induce EMT in intestinal epithelial cells 153. Anti-IL-17A treatment alleviated intestinal fibrosis by reducing EMT in mouse intestines 153. Consistent with these data, in a TNBS-induced intestinal fibrosis mouse model, treatment with anti-IL-17 antibody significantly alleviated intestinal fibrosis and reduced both mRNA and protein levels of collagen 3, TNF-α, TIMP-1, and MMP-2, as well as the decreased levels of profibrogenic cytokines IL-1β, TGF-β1, and TNF-α 154. However, secukinumab, a human anti-IL-17A monoclonal antibody, failed to show efficacy in CD and in fact led to worsening of intestinal inflammation in some patients, suggesting that baseline levels of IL-17 are necessary for intestinal homeostasis 155.
IL-4, IL-13 and the Th2 response.
IL-4 and IL-13 are prototypical Th2 cytokines. Although it was initially believed that Th1 cells are involved in CD whereas Th2 or Th2-like cells are associated with UC 156, Th2 immune responses have also been detected in late stages of experimental and human CD 157,158. Previous studies in several fibroblastic synovial cell lines indicated that IL-4 might be a pro-fibrogenic cytokine promoting collagen and fibronectin synthesis in normal healing and pathological fibrosis 159. In addition, the treatment of human hepatic fibroblasts with IL-4 increased collagen production 160. However, in IBD the role of IL-4 is still controversial. Although IL4 mRNA expression in the intestinal mucosa in both CD and UC patients is almost undetectable 161, anti-IL-4 administration leads to a striking amelioration of oxazolone colitis 162. IL-13 signals through IL-13Rα2 to activate the TGF-β1 promoter and the prevention of IL-13Rα2 expression reduced production of TGF-β1 in oxazolone- or TNBS-induced colitis 163,164. Additionally, IL-13 and its receptor were found to be overexpressed in areas of fibrosis in CD patients 165. However, anrukinzumab, an anti-interleukin 13 monoclonal antibody, showed no therapeutic effect in patients with active UC in a phase IIa randomized multicenter study 166. The effect of another IL-13-neutralizing antibody, tralokinumab, was evaluated in a randomized, double-blind, placebo-controlled, phase IIa study as add-on therapy in adults with moderate-to-severe UC despite standard treatments and the results demonstrated that did not significantly improve clinical response, although a higher than placebo clinical remission rate suggested that tralokinumab might benefit some UC patients 167.
IL-34.
IL-34 is a novel cytokine identified as a tissue-specific ligand of CSF-1 receptor (CSF-1R) 130. Although IL-34 shares the receptor CSF-1R with CSF1and M-CSF, IL-34 shows very little homology with them and IL-34 has also two distinct receptors (PTP-ζ) and CD138 (syndecan-1) 168. It belongs to the 4-helical cytokine family, which usually functionally mimics the PDGF and VEGF cystine knot dimers 130. In humans, IL-34 has a wide distribution in various tissues, including the heart, brain, lung, liver, kidney, spleen, thymus, testis, ovary, small intestine, prostate, colon, and spleen, and it can be expressed by synovial fibroblasts, immune, epithelial, endothelial, cancer cells and adipocytes 169. IL-34 induces lymphocyte differentiation, proliferation, and regulates the synthesis of inflammatory components and aberrant expression of IL‐34 in several autoimmune disorders, such as lupus, arthritis, systemic sclerosis 130. IL-34 is overexpressed in chronic hepatitis C liver fibrosis and induce profibrotic macrophages, and serum interleukin-34 level can be an indicator of liver fibrosis in patients with chronic hepatitis B, as well as a marker of liver fibrosis in patients with non-alcoholic fatty liver disease 170–172. IL-34 deficient mice exhibit less renal fibrosis during the chronic phase after ischemia/reperfusion injury 173. In the gut IL-34 is constitutively expressed in human small intestine and colon and shows a higher expression level in stricturing CDs compared to controls 174. It functions as a direct activator of intestinal fibroblasts, who respond with increased collagen expression, including COL1A1 and COL3A1, in a p38MAPK dependent manner 174. IL-34 knockdown results in decreased collagen production in CD fibroblasts isolated from CD strictures without affecting cell survival 174.
Host-microbiome interactions and intestinal fibrosis
The gut microbiota, a highly complex and dynamic population of microorganisms, has become the subject of intense investigation in recent years and plays a major role in human health and disease 175, and compositional and metabolic alterations of the gut microbiota (dysbiosis) are key factors involved in IBD pathogenesis 19. In individuals with a genetic susceptibility to IBD, microbial antigens and products induce functional changes of the intestinal mucosa and immune response which are linked to IBD onset and progression 176.
Innate immune response is the first line of defense against invading pathogens, and initiation of this response depends on the recognition of pathogen-associated molecular patterns (PAMPs) by a series of pathogen recognition receptors (PRRs) 177. PRRs are expressed by both intestinal immune (dendritic cells, macrophages, lymphocytes, etc.) and non-immune cells (epithelial cells, fibroblasts, endothelial cells, etc.) 178. Primary human intestinal myofibroblasts express multiple PRRs, including toll-like receptor (TLR) 1–9, as well as nucleotide-binding oligomerization domain-containing protein (NOD) 1 and NOD2 179. Lipopolysaccharide (LPS), predominantly derived from gram-negative bacteria and recognized by TLR4, promotes profibrotic activation of intestinal fibroblasts, with enhanced NF-κB promoter activity and increased collagen production 180. DSS-induced intestinal fibrosis is decreased in TLR4-deficient mice, with reduced TGF-β production by peritoneal macrophages and less collagen production 181. Flagellin, which potentiates the expression of the IL-33 receptor ST2 in the intestinal epithelium, promotes intestinal fibrosis in a TLR5- and NOD-like receptor family CARD domain-containing protein (NLRC) 4-dependent manner 117. Supporting the notion of microbial driven fibrogenesis, CD patients carrying NOD2 mutations display a fibrostenotic phenotype 182.
Bacteria-driven IBD animal models, microbes or microbial components are used to study intestinal fibrosis, such as AIEC infection and Salmonella enterica serovar Typhimurium (S. Typhimurium) infection mouse models 182,183. On one hand, research in these animal models provides evidence of the relationship between gut microbiome and intestinal fibrosis and, on the other hand, uncovers new molecules and mechanisms involved in intestinal fibrogenesis.
AIEC is a pathotype of E. coli which adheres to the gut epithelium and causes chronic intestinal inflammation in genetically susceptible hosts 184. AIEC strains are found more frequently in ileal specimens from CD patients and are suspected to be involved in the initiation and/or progression of local inflammation 185. In mice, chronic AIEC infection leads to tissue pathology in the small and large bowel of mice, especially in the cecum, with elevated Th1 and Th17 responses 186. In addition, cecum from AIEC-infected mice exhibits extensive ECM deposition, higher expression levels of collagen type I and III, and enhanced expression of profibrotic mediators, such as TGF-β1, connective-tissue growth factor (CTGF) and insulin-like growth factor I (IGF-I), all of which are also observed in CD patients 186. However, intestinal strictures do not occur in this model, and inflammation and fibrosis is mouse strain-dependent.
S. Typhimurium is a common gastrointestinal bacterial pathogen that causes food poisoning and gastroenteritis in millions of people each year. S. Typhimurium infection induces chronic Salmonella colonization of the murine cecum and colon, with severe transmural inflammation 117. S. Typhimurium infection also increases the susceptibility of DSS-treated and IL-10−/− mice to intestinal inflammation 187. Although in humans S. Typhimurium infection has not been implicated in the pathogenesis of CD or intestinal fibrosis, S. Typhimurium-infected mice exhibit enhanced production of TGF-β and IGF-I, along with extensive collagen I deposition in the cecal mucosa, submucosa, and muscularis mucosa 187 and large numbers of fibroblasts and myofibroblasts in fibrotic areas 188. A modified Salmonella-induced intestinal fibrosis model has been reported and was used to detect the role of flagellin and IL-33 in intestinal fibrosis 117. In this model, AIEC and an attenuated strain of S. Typhimurium were co-colonized and induced intestinal fibrosis in a flagellin-dependent manner with the activation of IL-33-ST2 signaling 117. No animal model can fully mimic human IBD fibrosis, but deeper investigation of the mechanism of IBD and further characterization of novel animal models will support the interrogation of fibrotic mechanisms in vivo.
Creeping fat and smooth muscle hyperplasia in intestinal fibrosis
Fat wrapping, also called creeping fat, a pathologically altered mesenteric fat tissue adjacent to CD-involved intestinal segments and was described almost 100 years ago, but its functional implications have only recently received attention 189. Creeping fat is highly linked to stricturing disease and its extent correlates closely with the degree of transmural inflammation 190, a clinically relevant observation. When the mesentery is removed extensively for ileocolic resections (as opposed to minimal mesenteric resection) CD recurrence is reduced, and fewer reoperations are required 43. The mechanism underlying the formation of creeping fat remains unclear. One factor essential to the formation of mesenteric adipose tissue in CD is adipocyte hyperplasia, which occurs by recruitment and differentiation of adipose tissue-derived stem cells (ASCs). CD ASCs have elevated proliferative, invasive and phagocytic capacities, which may contribute to the creeping fat development 191. In CD ASCs exhibit a unique DNA methylation and gene expression profile 192 and CD creeping and mesenteric fat shows a microbiome signature that is absent in subcutaneous fat 193. Immune-non-immune cell interactions are critical to the function of creeping fat. Compared with tissue from control subjects, both T and B memory cells are increased within the creeping fat of CD patients 194, and CD mesenteric tissue contains greater amounts of adipocyte-derived chemokines which may actively recruit T and B lymphocytes 194. Of critical relevance, CD creeping fat is associated with muscularis propria hyperplasia, which is now believed to be the major factor contributing to the luminal narrowing in gut stenosis (Figure 2) 189,195. Our own preliminary observations suggest a novel role of creeping fat derived fatty acids on human intestinal muscle cells (HIMCs) hyperplasia, which contributes to the intestinal stricture formation 196,197. Exposure of HIMC to whole creeping fat tissue and fat-conditioned medium dramatically upregulates HIMC proliferation compared with UC and normal mesenteric fat 197. Creeping fat-derived mediators such as free fatty acids, but not adipokines, increase a differential and selective proliferative response by HIMC, which is dependent on p38MAPK, PKC, and PI3K 196,197. In addition, creeping fat may be linked with gut wall fibrosis. The adipocyte-dependent microenvironment within the creeping fat of patients with CD has been reported to promote an M2 macrophage subtype with secretion of large amounts of pro-fibrotic factors such as TGF-β, leading to intestinal fibrosis 198.
Pathway to develop anti-fibrotic trials in IBD
Despite the significant advances in the field of fibrosis, including features unique to the intestine such as creeping fat and microbial influence, trials testing drugs with an anti-fibrotic mechanism have yet to be started in IBD. This is largely due to the major obstacle of missing consensus on definitions and the absence of clinical endpoints. While multiple biomarkers have been proposed to stratify patients without any complications at diagnosis into at-risk populations for future strictures, none of the current markers are able to discriminate future stricturing from penetrating behavior with high enough accuracy to allow enrolling CD patients into fibrosis prevention trials 199,200. In addition, at present no accurate biomarkers or imaging technique can accurately quantify fibrosis in a stricture, which make studies relying solely on ECM changes as endpoints challenging. Cross-sectional imaging techniques, including ultrasound, computed tomography enterography and magnetic resonance enterography can detect intestinal strictures or assess inflammation with high accuracy but cannot determine fibrosis degree 201, making it practically impossible to assess responses to anti-fibrotic therapy. One possible starting point is to choose a population with existing strictures, in which resection studies have provided an assessment of the amount of present fibrosis, muscle thickening and inflammation 202. In these patients a background therapy with anti-inflammatory drugs can lead to a mitigation in obstructive symptoms that then allows the randomization of patients to an anti-fibrotic therapy versus placebo. Clinically meaningful endpoints could be absence of obstructive symptoms, stricture morphology changes on cross sectional imaging or reduced number of endoscopic or surgical interventions.
To accomplish translation of novel anti-fibrotics the field of stricturing IBD is in dire need of reliable definitions. Systematic reviews reveal tremendous heterogeneity in the definitions for stricturing IBD on endoscopy and cross-sectional imaging 201. Recently, the global consortium Stenosis Therapy and Anti-Fibrotic Therapy (STAR) created clear definitions for what defines a stricture and what constitutes improvement 203. Multiple projects are now underway to build the monitoring tools and endpoints for clinical trials in fibrostenosing IBD, including a patient reported outcome tool, a stricture radiology index and dynamic fibrosis turnover markers.
Finally, successful development of markers to predict or quantify fibrosis in the future will be based on development of a solid histopathologic gold standard. Typically, candidate markers are evaluated in patients with symptomatic strictures, who then undergo imaging, followed by bowel resection with grading of the intestinal tissue for inflammation and fibrosis 204–211. Next, construct validity is determined by correlating histologic evaluations of the degree of fibrosis and/or inflammation on the resected tissue to imaging results. Multiple histopathology indices have been proposed, but none has been validated following modern methodological standards 212, and their evaluation is highly variable across indices 213. A reliable, validated and responsive histopathologic standard would deliver the needed discriminatory ability as a basis to optimize and compare cross sectional imaging techniques. An international working group comprised of IBD pathologists and gastroenterologists under the umbrella of the STAR consortium is working on building a validated histopathology index for stricturing IBD. Ultimate progress will be fueled by combining mechanistic preclinical studies with the newly created endpoints in state of-the-art drug development programs (Figure 3).
Acknowledgements
Funding:
This work was supported by the Helmsley Charitable Trust through the Stenosis Therapy and Anti-Fibrotic Research (STAR) Consortium (No. 3081 to F.R.), the Crohn’s and Colitis Foundation (No. 569125 to F.R.), the National Institute of Health (K08DK110415 and R01DK123233 to F.R.; R01HL111314 and R01DK123233 to D.V.W, P30DK097948 and T32DK083251 to C.F., R01DK120679, P01HL147823, P50AA024333, U01AA026938 to J.M.B.), American Heart Association (18SFRN34170442 to D.V.W.), the Kenneth Rainin Foundation to F.R. and the Cleveland Clinic through the LabCo program to F.R.
Footnotes
Competing interests:
F.R. is consultant to Agomab, Allergan, AbbVie, Boehringer-Ingelheim, Celgene, Cowen, Genentech, Gilead, Gossamer, Guidepoint, Helmsley, Index Pharma, Jannsen, Koutif, Metacrine, Morphic, Pfizer, Pliant, Prometheus Biosciences, Receptos, RedX, Roche, Samsung, Takeda, Techlab, Thetis, UCB. None of the other authors has any competing interest to declare.
References
- 1.Ng SC, Shi HY, Hamidi N, et al. Worldwide incidence and prevalence of inflammatory bowel disease in the 21st century: a systematic review of population-based studies. Lancet. 2018;390(10114):2769–2778. [DOI] [PubMed] [Google Scholar]
- 2.Neurath MF. Targeting immune cell circuits and trafficking in inflammatory bowel disease. Nat Immunol. 2019;20(8):970–979. [DOI] [PubMed] [Google Scholar]
- 3.Yoo JH, Holubar S, Rieder F. Fibrostenotic strictures in Crohn’s disease. Intest Res. 2020;18(4):379–401. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Li J, Mao R, Kurada S, et al. Pathogenesis of fibrostenosing Crohn’s disease. Transl Res. 2019;209:39–54. [DOI] [PubMed] [Google Scholar]
- 5.Stenke E, Bourke B, Knaus U. Crohn’s Strictures-Moving Away from the Knife. Front Pediatr. 2017;5:141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Thia KT, Sandborn WJ, Harmsen WS, Zinsmeister AR, Loftus EV Jr. Risk factors associated with progression to intestinal complications of Crohn’s disease in a population-based cohort. Gastroenterology. 2010;139(4):1147–1155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Latella G, Rieder F. Time to Look Underneath the Surface: Ulcerative Colitis-Associated Fibrosis. J Crohns Colitis. 2015;9(11):941–942. [DOI] [PubMed] [Google Scholar]
- 8.Gordon IO, Agrawal N, Willis E, et al. Fibrosis in ulcerative colitis is directly linked to severity and chronicity of mucosal inflammation. Aliment Pharmacol Ther. 2018;47(7):922–939. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Gundersen MD, Goll R, Fenton CG, et al. Fibrosis Mediators in the Colonic Mucosa of Acute and Healed Ulcerative Colitis. Clin Transl Gastroenterol. 2019;10(10):e00082. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Rieder F, Kessler S, Sans M, Fiocchi C. Animal models of intestinal fibrosis: new tools for the understanding of pathogenesis and therapy of human disease. Am J Physiol Gastrointest Liver Physiol. 2012;303(7):G786–801. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Cohen LJ, Cho JH, Gevers D, Chu H. Genetic Factors and the Intestinal Microbiome Guide Development of Microbe-Based Therapies for Inflammatory Bowel Diseases. Gastroenterology. 2019;156(8):2174–2189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Hall AB, Tolonen AC, Xavier RJ. Human genetic variation and the gut microbiome in disease. Nat Rev Genet. 2017;18(11):690–699. [DOI] [PubMed] [Google Scholar]
- 13.Luca F, Kupfer SS, Knights D, Khoruts A, Blekhman R. Functional Genomics of Host-Microbiome Interactions in Humans. Trends Genet. 2018;34(1):30–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Khor B, Gardet A, Xavier RJ. Genetics and pathogenesis of inflammatory bowel disease. Nature. 2011;474(7351):307–317. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Cleynen I, Boucher G, Jostins L, et al. Inherited determinants of Crohn’s disease and ulcerative colitis phenotypes: a genetic association study. Lancet. 2016;387(10014):156–167. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Jostins L, Ripke S, Weersma RK, et al. Host-microbe interactions have shaped the genetic architecture of inflammatory bowel disease. Nature. 2012;491(7422):119–124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Parkes M The genetics universe of Crohn’s disease and ulcerative colitis. Dig Dis. 2012;30 Suppl 1:78–81. [DOI] [PubMed] [Google Scholar]
- 18.Abegunde AT, Muhammad BH, Bhatti O, Ali T. Environmental risk factors for inflammatory bowel diseases: Evidence based literature review. World J Gastroenterol. 2016;22(27):6296–6317. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Ni J, Wu GD, Albenberg L, Tomov VT. Gut microbiota and IBD: causation or correlation? Nat Rev Gastroenterol Hepatol. 2017;14(10):573–584. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Davies JM, Abreu MT. The innate immune system and inflammatory bowel disease. Scand J Gastroenterol. 2015;50(1):24–33. [DOI] [PubMed] [Google Scholar]
- 21.Geremia A, Biancheri P, Allan P, Corazza GR, Di Sabatino A. Innate and adaptive immunity in inflammatory bowel disease. Autoimmun Rev. 2014;13(1):3–10. [DOI] [PubMed] [Google Scholar]
- 22.Huang Y, Chen Z. Inflammatory bowel disease related innate immunity and adaptive immunity. Am J Transl Res. 2016;8(6):2490–2497. [PMC free article] [PubMed] [Google Scholar]
- 23.de Souza HS, Fiocchi C. Immunopathogenesis of IBD: current state of the art. Nat Rev Gastroenterol Hepatol. 2016;13(1):13–27. [DOI] [PubMed] [Google Scholar]
- 24.Wynn TA. Cellular and molecular mechanisms of fibrosis. J Pathol. 2008;214(2):199–210. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Lee SB, Kalluri R. Mechanistic connection between inflammation and fibrosis. Kidney Int Suppl. 2010(119):S22–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Mack M. Inflammation and fibrosis. Matrix Biol. 2018;68–69:106–121. [DOI] [PubMed] [Google Scholar]
- 27.Prockop DJ. Inflammation, fibrosis, and modulation of the process by mesenchymal stem/stromal cells. Matrix Biol. 2016;51:7–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Latella G, Di Gregorio J, Flati V, Rieder F, Lawrance IC. Mechanisms of initiation and progression of intestinal fibrosis in IBD. Scand J Gastroenterol. 2015;50(1):53–65. [DOI] [PubMed] [Google Scholar]
- 29.Arpino V, Brock M, Gill SE. The role of TIMPs in regulation of extracellular matrix proteolysis. Matrix Biol. 2015;44–46:247–254. [DOI] [PubMed] [Google Scholar]
- 30.Thomson CA, Nibbs RJ, McCoy KD, Mowat AM. Immunological roles of intestinal mesenchymal cells. Immunology. 2020;160(4):313–324. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Koliaraki V, Pallangyo CK, Greten FR, Kollias G. Mesenchymal Cells in Colon Cancer. Gastroenterology. 2017;152(5):964–979. [DOI] [PubMed] [Google Scholar]
- 32.Powell DW, Pinchuk IV, Saada JI, Chen X, Mifflin RC. Mesenchymal cells of the intestinal lamina propria. Annu Rev Physiol. 2011;73:213–237. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Roulis M, Flavell RA. Fibroblasts and myofibroblasts of the intestinal lamina propria in physiology and disease. Differentiation. 2016;92(3):116–131. [DOI] [PubMed] [Google Scholar]
- 34.Rieder F, Fiocchi C. Intestinal fibrosis in IBD--a dynamic, multifactorial process. Nat Rev Gastroenterol Hepatol. 2009;6(4):228–235. [DOI] [PubMed] [Google Scholar]
- 35.Koumas L, Smith TJ, Feldon S, Blumberg N, Phipps RP. Thy-1 expression in human fibroblast subsets defines myofibroblastic or lipofibroblastic phenotypes. Am J Pathol. 2003;163(4):1291–1300. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Speca S, Giusti I, Rieder F, Latella G. Cellular and molecular mechanisms of intestinal fibrosis. World J Gastroenterol. 2012;18(28):3635–3661. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Chen W, Lu C, Hirota C, Iacucci M, Ghosh S, Gui X. Smooth Muscle Hyperplasia/Hypertrophy is the Most Prominent Histological Change in Crohn’s Fibrostenosing Bowel Strictures: A Semiquantitative Analysis by Using a Novel Histological Grading Scheme. J Crohns Colitis. 2017;11(1):92–104. [DOI] [PubMed] [Google Scholar]
- 38.Valatas V, Filidou E, Drygiannakis I, Kolios G. Stromal and immune cells in gut fibrosis: the myofibroblast and the scarface. Ann Gastroenterol. 2017;30(4):393–404. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Rieder F, Brenmoehl J, Leeb S, Scholmerich J, Rogler G. Wound healing and fibrosis in intestinal disease. Gut. 2007;56(1):130–139. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Sahebally SM, Burke JP, Chang KH, Kiernan MG, O’Connell PR, Coffey JC. Circulating fibrocytes and Crohn’s disease. Br J Surg. 2013;100(12):1549–1556. [DOI] [PubMed] [Google Scholar]
- 41.Fibrocytes Sorrentino D., inflammation, and fibrosis in Crohn’s disease: another piece of the puzzle. Dig Dis Sci. 2014;59(4):699–701. [DOI] [PubMed] [Google Scholar]
- 42.Sazuka S, Katsuno T, Nakagawa T, et al. Fibrocytes are involved in inflammation as well as fibrosis in the pathogenesis of Crohn’s disease. Dig Dis Sci. 2014;59(4):760–768. [DOI] [PubMed] [Google Scholar]
- 43.Coffey CJ, Kiernan MG, Sahebally SM, et al. Inclusion of the Mesentery in Ileocolic Resection for Crohn’s Disease is Associated With Reduced Surgical Recurrence. J Crohns Colitis. 2018;12(10):1139–1150. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Kiernan MG, Coffey JC, Sahebally SM, et al. Systemic Molecular Mediators of Inflammation Differentiate Between Crohn’s Disease and Ulcerative Colitis, Implicating Threshold Levels of IL-10 and Relative Ratios of Pro-inflammatory Cytokines in Therapy. J Crohns Colitis. 2020;14(1):118–129. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Li C, Kuemmerle JF. The fate of myofibroblasts during the development of fibrosis in Crohn’s disease. J Dig Dis. 2020;21(6):326–331. [DOI] [PubMed] [Google Scholar]
- 46.Rieder F, Fiocchi C. Intestinal fibrosis in inflammatory bowel disease - Current knowledge and future perspectives. J Crohns Colitis. 2008;2(4):279–290. [DOI] [PubMed] [Google Scholar]
- 47.Ippolito C, Colucci R, Segnani C, et al. Fibrotic and Vascular Remodelling of Colonic Wall in Patients with Active Ulcerative Colitis. J Crohns Colitis. 2016;10(10):1194–1204. [DOI] [PubMed] [Google Scholar]
- 48.Lamouille S, Xu J, Derynck R. Molecular mechanisms of epithelial-mesenchymal transition. Nat Rev Mol Cell Biol. 2014;15(3):178–196. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Kalluri R, Weinberg RA. The basics of epithelial-mesenchymal transition. J Clin Invest. 2009;119(6):1420–1428. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Lovisa S, Genovese G, Danese S. Role of Epithelial-to-Mesenchymal Transition in Inflammatory Bowel Disease. J Crohns Colitis. 2019;13(5):659–668. [DOI] [PubMed] [Google Scholar]
- 51.Mehta SJ, Lewis A, Nijhuis A, et al. Epithelial down-regulation of the miR-200 family in fibrostenosing Crohn’s disease is associated with features of epithelial to mesenchymal transition. J Cell Mol Med. 2018;22(11):5617–5628. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Ortiz-Masia D, Salvador P, Macias-Ceja DC, et al. WNT2b Activates Epithelial-mesenchymal Transition Through FZD4: Relevance in Penetrating Crohn s Disease. J Crohns Colitis. 2020;14(2):230–239. [DOI] [PubMed] [Google Scholar]
- 53.Dejana E, Hirschi KK, Simons M. The molecular basis of endothelial cell plasticity. Nat Commun. 2017;8:14361. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Rieder F, Kessler SP, West GA, et al. Inflammation-induced endothelial-to-mesenchymal transition: a novel mechanism of intestinal fibrosis. Am J Pathol. 2011;179(5):2660–2673. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Mintet E, Rannou E, Buard V, et al. Identification of Endothelial-to-Mesenchymal Transition as a Potential Participant in Radiation Proctitis. Am J Pathol. 2015;185(9):2550–2562. [DOI] [PubMed] [Google Scholar]
- 56.Dobie R, Henderson NC. Unravelling fibrosis using single-cell transcriptomics. Curr Opin Pharmacol. 2019;49:71–75. [DOI] [PubMed] [Google Scholar]
- 57.Xie T, Wang Y, Deng N, et al. Single-Cell Deconvolution of Fibroblast Heterogeneity in Mouse Pulmonary Fibrosis. Cell Rep. 2018;22(13):3625–3640. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Nabhan AN, Brownfield DG, Harbury PB, Krasnow MA, Desai TJ. Single-cell Wnt signaling niches maintain stemness of alveolar type 2 cells. Science. 2018;359(6380):1118–1123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Mederacke I, Hsu CC, Troeger JS, et al. Fate tracing reveals hepatic stellate cells as dominant contributors to liver fibrosis independent of its aetiology. Nat Commun. 2013;4:2823. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Iwaisako K, Jiang C, Zhang M, et al. Origin of myofibroblasts in the fibrotic liver in mice. Proc Natl Acad Sci U S A. 2014;111(32):E3297–3305. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Krenkel O, Hundertmark J, Ritz TP, Weiskirchen R, Tacke F. Single Cell RNA Sequencing Identifies Subsets of Hepatic Stellate Cells and Myofibroblasts in Liver Fibrosis. Cells. 2019;8(5). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Ramachandran P, Dobie R, Wilson-Kanamori JR, et al. Resolving the fibrotic niche of human liver cirrhosis at single-cell level. Nature. 2019;575(7783):512–518. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Dobie R, Wilson-Kanamori JR, Henderson BEP, et al. Single-Cell Transcriptomics Uncovers Zonation of Function in the Mesenchyme during Liver Fibrosis. Cell Rep. 2019;29(7):1832–1847 e1838. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Kramann R, Machado F, Wu H, et al. Parabiosis and single-cell RNA sequencing reveal a limited contribution of monocytes to myofibroblasts in kidney fibrosis. JCI Insight. 2018;3(9). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Wu H, Kirita Y, Donnelly EL, Humphreys BD. Advantages of Single-Nucleus over Single-Cell RNA Sequencing of Adult Kidney: Rare Cell Types and Novel Cell States Revealed in Fibrosis. J Am Soc Nephrol. 2019;30(1):23–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Fu H, Tian Y, Zhou L, et al. Tenascin-C Is a Major Component of the Fibrogenic Niche in Kidney Fibrosis. J Am Soc Nephrol. 2017;28(3):785–801. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Kinchen J, Chen HH, Parikh K, et al. Structural Remodeling of the Human Colonic Mesenchyme in Inflammatory Bowel Disease. Cell. 2018;175(2):372–386e317. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Martin JC, Chang C, Boschetti G, et al. Single-Cell Analysis of Crohn’s Disease Lesions Identifies a Pathogenic Cellular Module Associated with Resistance to Anti-TNF Therapy. Cell. 2019;178(6):1493–1508 e1420. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Tsukui T, Sun KH, Wetter JB, et al. Collagen-producing lung cell atlas identifies multiple subsets with distinct localization and relevance to fibrosis. Nat Commun. 2020;11(1):1920. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Friedrich M, Pohin M, Powrie F. Cytokine Networks in the Pathophysiology of Inflammatory Bowel Disease. Immunity. 2019;50(4):992–1006. [DOI] [PubMed] [Google Scholar]
- 71.Graham MF, Bryson GR, Diegelmann RF. Transforming growth factor beta 1 selectively augments collagen synthesis by human intestinal smooth muscle cells. Gastroenterology. 1990;99(2):447–453. [DOI] [PubMed] [Google Scholar]
- 72.di Mola FF, Friess H, Scheuren A, et al. Transforming growth factor-betas and their signaling receptors are coexpressed in Crohn’s disease. Ann Surg. 1999;229(1):67–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Di Sabatino A, Jackson CL, Pickard KM, et al. Transforming growth factor beta signalling and matrix metalloproteinases in the mucosa overlying Crohn’s disease strictures. Gut. 2009;58(6):777–789. [DOI] [PubMed] [Google Scholar]
- 74.Ihara S, Hirata Y, Koike K. TGF-beta in inflammatory bowel disease: a key regulator of immune cells, epithelium, and the intestinal microbiota. J Gastroenterol. 2017;52(7):777–787. [DOI] [PubMed] [Google Scholar]
- 75.Miyazawa K, Miyazono K. Regulation of TGF-beta Family Signaling by Inhibitory Smads. Cold Spring Harb Perspect Biol. 2017;9(3). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Zhang YE. Non-Smad pathways in TGF-beta signaling. Cell Res. 2009;19(1):128–139. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Drygiannakis I, Valatas V, Sfakianaki O, et al. Proinflammatory cytokines induce crosstalk between colonic epithelial cells and subepithelial myofibroblasts: implication in intestinal fibrosis. J Crohns Colitis. 2013;7(4):286–300. [DOI] [PubMed] [Google Scholar]
- 78.Leeb SN, Vogl D, Falk W, Scholmerich J, Rogler G, Gelbmann CM. Regulation of migration of human colonic myofibroblasts. Growth Factors. 2002;20(2):81–91. [DOI] [PubMed] [Google Scholar]
- 79.Rodansky ES, Johnson LA, Huang S, Spence JR, Higgins PD. Intestinal organoids: a model of intestinal fibrosis for evaluating anti-fibrotic drugs. Exp Mol Pathol. 2015;98(3):346–351. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Ma Y, Guan Q, Bai A, et al. Targeting TGF-beta1 by employing a vaccine ameliorates fibrosis in a mouse model of chronic colitis. Inflamm Bowel Dis. 2010;16(6):1040–1050. [DOI] [PubMed] [Google Scholar]
- 81.Izzo R, Bevivino G, De Simone V, et al. Knockdown of Smad7 With a Specific Antisense Oligonucleotide Attenuates Colitis and Colitis-Driven Colonic Fibrosis in Mice. Inflamm Bowel Dis. 2018;24(6):1213–1224. [DOI] [PubMed] [Google Scholar]
- 82.Li C, Grider JR, Murthy KS, et al. Endoplasmic Reticulum Stress in Subepithelial Myofibroblasts Increases the TGF-beta1 Activity That Regulates Fibrosis in Crohn’s Disease. Inflamm Bowel Dis. 2020;26(6):809–819. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Fuchs PO, Calitz C, Pavlovic N, et al. Fibrin fragment E potentiates TGF-beta-induced myofibroblast activation and recruitment. Cell Signal. 2020;72:109661. [DOI] [PubMed] [Google Scholar]
- 84.Franze E, Monteleone I, Laudisi F, et al. Cadherin-11 Is a Regulator of Intestinal Fibrosis. J Crohns Colitis. 2020;14(3):406–417. [DOI] [PubMed] [Google Scholar]
- 85.Steiner CA, Rodansky ES, Johnson LA, et al. AXL Is a Potential Target for the Treatment of Intestinal Fibrosis. Inflamm Bowel Dis. 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Guan Y, Tan Y, Liu W, et al. NF-E2-Related Factor 2 Suppresses Intestinal Fibrosis by Inhibiting Reactive Oxygen Species-Dependent TGF-beta1/SMADs Pathway. Dig Dis Sci. 2018;63(2):366–380. [DOI] [PubMed] [Google Scholar]
- 87.Dinarello CA. Overview of the IL-1 family in innate inflammation and acquired immunity. Immunol Rev. 2018;281(1):8–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Borthwick LA. The IL-1 cytokine family and its role in inflammation and fibrosis in the lung. Semin Immunopathol. 2016;38(4):517–534. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Bersudsky M, Luski L, Fishman D, et al. Non-redundant properties of IL-1alpha and IL-1beta during acute colon inflammation in mice. Gut. 2014;63(4):598–609. [DOI] [PubMed] [Google Scholar]
- 90.Scarpa M, Kessler S, Sadler T, et al. The epithelial danger signal IL-1alpha is a potent activator of fibroblasts and reactivator of intestinal inflammation. Am J Pathol. 2015;185(6):1624–1637. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Suwara MI, Green NJ, Borthwick LA, et al. IL-1alpha released from damaged epithelial cells is sufficient and essential to trigger inflammatory responses in human lung fibroblasts. Mucosal Immunol. 2014;7(3):684–693. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Doerner AM, Zuraw BL. TGF-beta1 induced epithelial to mesenchymal transition (EMT) in human bronchial epithelial cells is enhanced by IL-1beta but not abrogated by corticosteroids. Respir Res. 2009;10:100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Kolb M, Margetts PJ, Anthony DC, Pitossi F, Gauldie J. Transient expression of IL-1beta induces acute lung injury and chronic repair leading to pulmonary fibrosis. J Clin Invest. 2001;107(12):1529–1536. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Gasse P, Mary C, Guenon I, et al. IL-1R1/MyD88 signaling and the inflammasome are essential in pulmonary inflammation and fibrosis in mice. J Clin Invest. 2007;117(12):3786–3799. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Wilson MS, Madala SK, Ramalingam TR, et al. Bleomycin and IL-1beta-mediated pulmonary fibrosis is IL-17A dependent. J Exp Med. 2010;207(3):535–552. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Reiter FP, Wimmer R, Wottke L, et al. Role of interleukin-1 and its antagonism of hepatic stellate cell proliferation and liver fibrosis in the Abcb4(−/−) mouse model. World J Hepatol. 2016;8(8):401–410. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Lemos DR, McMurdo M, Karaca G, et al. Interleukin-1beta Activates a MYC-Dependent Metabolic Switch in Kidney Stromal Cells Necessary for Progressive Tubulointerstitial Fibrosis. J Am Soc Nephrol. 2018;29(6):1690–1705. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Okuno T, Andoh A, Bamba S, et al. Interleukin-1beta and tumor necrosis factor-alpha induce chemokine and matrix metalloproteinase gene expression in human colonic subepithelial myofibroblasts. Scand J Gastroenterol. 2002;37(3):317–324. [DOI] [PubMed] [Google Scholar]
- 99.Graham MF, Willey A, Adams J, Yager D, Diegelmann RF. Interleukin 1 beta down-regulates collagen and augments collagenase expression in human intestinal smooth muscle cells. Gastroenterology. 1996;110(2):344–350. [DOI] [PubMed] [Google Scholar]
- 100.Graham MF, Willey A, Zhu YN, Yager DR, Sugerman HJ, Diegelmann RF. Corticosteroids repress the interleukin 1 beta-induced secretion of collagenase in human intestinal smooth muscle cells. Gastroenterology. 1997;113(6):1924–1929. [DOI] [PubMed] [Google Scholar]
- 101.Buhl AL, Wenzel J. Interleukin-36 in Infectious and Inflammatory Skin Diseases. Front Immunol. 2019;10:1162. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Yuan ZC, Xu WD, Liu XY, Liu XY, Huang AF, Su LC. Biology of IL-36 Signaling and Its Role in Systemic Inflammatory Diseases. Front Immunol. 2019;10:2532. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Nishida A, Inatomi O, Fujimoto T, Imaeda H, Tani M, Andoh A. Interleukin-36alpha Induces Inflammatory Mediators From Human Pancreatic Myofibroblasts Via a MyD88 Dependent Pathway. Pancreas. 2017;46(4):539–548. [DOI] [PubMed] [Google Scholar]
- 104.Murrieta-Coxca JM, Rodriguez-Martinez S, Cancino-Diaz ME, Markert UR, Favaro RR, Morales-Prieto DM. IL-36 Cytokines: Regulators of Inflammatory Responses and Their Emerging Role in Immunology of Reproduction. Int J Mol Sci. 2019;20(7). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Chen F, Qu M, Zhang F, et al. IL-36 s in the colorectal cancer: is interleukin 36 good or bad for the development of colorectal cancer? BMC Cancer. 2020;20(1):92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Scheibe K, Backert I, Wirtz S, et al. IL-36R signalling activates intestinal epithelial cells and fibroblasts and promotes mucosal healing in vivo. Gut. 2017;66(5):823–838. [DOI] [PubMed] [Google Scholar]
- 107.Russell SE, Horan RM, Stefanska AM, et al. IL-36alpha expression is elevated in ulcerative colitis and promotes colonic inflammation. Mucosal Immunol. 2016;9(5):1193–1204. [DOI] [PubMed] [Google Scholar]
- 108.Nishida A, Hidaka K, Kanda T, et al. Increased Expression of Interleukin-36, a Member of the Interleukin-1 Cytokine Family, in Inflammatory Bowel Disease. Inflamm Bowel Dis. 2016;22(2):303–314. [DOI] [PubMed] [Google Scholar]
- 109.Elias M, Zhao S, Le HT, et al. IL-36 in chronic inflammation and fibrosis - bridging the gap? J Clin Invest. 2021;131(2). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Boutet MA, Bart G, Penhoat M, et al. Distinct expression of interleukin (IL)-36alpha, beta and gamma, their antagonist IL-36Ra and IL-38 in psoriasis, rheumatoid arthritis and Crohn’s disease. Clin Exp Immunol. 2016;184(2):159–173. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Chi HH, Hua KF, Lin YC, et al. IL-36 Signaling Facilitates Activation of the NLRP3 Inflammasome and IL-23/IL-17 Axis in Renal Inflammation and Fibrosis. J Am Soc Nephrol. 2017;28(7):2022–2037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Scheibe K, Kersten C, Schmied A, et al. Inhibiting Interleukin 36 Receptor Signaling Reduces Fibrosis in Mice With Chronic Intestinal Inflammation. Gastroenterology. 2019;156(4):1082–1097 e1011. [DOI] [PubMed] [Google Scholar]
- 113.Lopetuso LR, Scaldaferri F, Pizarro TT. Emerging role of the interleukin (IL)-33/ST2 axis in gut mucosal wound healing and fibrosis. Fibrogenesis Tissue Repair. 2012;5(1):18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Kotsiou OS, Gourgoulianis KI, Zarogiannis SG. IL-33/ST2 Axis in Organ Fibrosis. Front Immunol. 2018;9:2432. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Beltran CJ, Nunez LE, Diaz-Jimenez D, et al. Characterization of the novel ST2/IL-33 system in patients with inflammatory bowel disease. Inflamm Bowel Dis. 2010;16(7):1097–1107. [DOI] [PubMed] [Google Scholar]
- 116.Kobori A, Yagi Y, Imaeda H, et al. Interleukin-33 expression is specifically enhanced in inflamed mucosa of ulcerative colitis. J Gastroenterol. 2010;45(10):999–1007. [DOI] [PubMed] [Google Scholar]
- 117.Imai J, Kitamoto S, Sugihara K, et al. Flagellin-mediated activation of IL-33-ST2 signaling by a pathobiont promotes intestinal fibrosis. Mucosal Immunol. 2019;12(3):632–643. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Zhao S, Dejanovic D, Yao P, et al. Selective deletion of MyD88 signaling in alpha-SMA positive cells ameliorates experimental intestinal fibrosis via post-transcriptional regulation. Mucosal Immunol. 2020;13(4):665–678. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Rose-John S Interleukin-6 Family Cytokines. Cold Spring Harb Perspect Biol. 2018;10(2). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Mudter J, Neurath MF. Il-6 signaling in inflammatory bowel disease: pathophysiological role and clinical relevance. Inflamm Bowel Dis. 2007;13(8):1016–1023. [DOI] [PubMed] [Google Scholar]
- 121.Papiris SA, Tomos IP, Karakatsani A, et al. High levels of IL-6 and IL-8 characterize early-on idiopathic pulmonary fibrosis acute exacerbations. Cytokine. 2018;102:168–172. [DOI] [PubMed] [Google Scholar]
- 122.Xiang DM, Sun W, Ning BF, et al. The HLF/IL-6/STAT3 feedforward circuit drives hepatic stellate cell activation to promote liver fibrosis. Gut. 2018;67(9):1704–1715. [DOI] [PubMed] [Google Scholar]
- 123.Li C, Kuemmerle JF. Mechanisms that mediate the development of fibrosis in patients with Crohn’s disease. Inflamm Bowel Dis. 2014;20(7):1250–1258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Li C, Iness A, Yoon J, et al. Noncanonical STAT3 activation regulates excess TGF-beta1 and collagen I expression in muscle of stricturing Crohn’s disease. J Immunol. 2015;194(7):3422–3431. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Lim WW, Ng B, Widjaja A, et al. Transgenic interleukin 11 expression causes cross-tissue fibro-inflammation and an inflammatory bowel phenotype in mice. PLoS One. 2020;15(1):e0227505. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Taga T, Kishimoto T. Gp130 and the interleukin-6 family of cytokines. Annu Rev Immunol. 1997;15:797–819. [DOI] [PubMed] [Google Scholar]
- 127.Schafer S, Viswanathan S, Widjaja AA, et al. IL-11 is a crucial determinant of cardiovascular fibrosis. Nature. 2017;552(7683):110–115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Widjaja AA, Singh BK, Adami E, et al. Inhibiting Interleukin 11 Signaling Reduces Hepatocyte Death and Liver Fibrosis, Inflammation, and Steatosis in Mouse Models of Nonalcoholic Steatohepatitis. Gastroenterology. 2019;157(3):777–792 e714. [DOI] [PubMed] [Google Scholar]
- 129.Cook SA, Schafer S. Hiding in Plain Sight: Interleukin-11 Emerges as a Master Regulator of Fibrosis, Tissue Integrity, and Stromal Inflammation. Annu Rev Med. 2020;71:263–276. [DOI] [PubMed] [Google Scholar]
- 130.Xu WD, Huang AF, Fu L, Liu XY, Su LC. Targeting IL-34 in inflammatory autoimmune diseases. J Cell Physiol. 2019;234(12):21810–21816. [DOI] [PubMed] [Google Scholar]
- 131.Curciarello R, Docena GH, MacDonald TT. The Role of Cytokines in the Fibrotic Responses in Crohn’s Disease. Front Med (Lausanne). 2017;4:126. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Veny M, Garrido-Trigo A, Corraliza AM, et al. Dissecting common and unique effects of anti-alpha4beta7 and anti-tumor necrosis factor treatment in ulcerative colitis. J Crohns Colitis. 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Frei R, Fournier N, Zeitz J, et al. Early Initiation of Anti-TNF is Associated with Favourable Long-term Outcome in Crohn’s Disease: 10-Year-Follow-up Data from the Swiss IBD Cohort Study. J Crohns Colitis. 2019;13(10):1292–1301. [DOI] [PubMed] [Google Scholar]
- 134.Di Sabatino A, Pender SL, Jackson CL, et al. Functional modulation of Crohn’s disease myofibroblasts by anti-tumor necrosis factor antibodies. Gastroenterology. 2007;133(1):137–149. [DOI] [PubMed] [Google Scholar]
- 135.Adler J, Rahal K, Swanson SD, et al. Anti-tumor necrosis factor alpha prevents bowel fibrosis assessed by messenger RNA, histology, and magnetization transfer MRI in rats with Crohn’s disease. Inflamm Bowel Dis. 2013;19(4):683–690. [DOI] [PubMed] [Google Scholar]
- 136.Pallotta N, Barberani F, Hassan NA, Guagnozzi D, Vincoli G, Corazziari E. Effect of infliximab on small bowel stenoses in patients with Crohn’s disease. World J Gastroenterol. 2008;14(12):1885–1890. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Kugathasan S, Denson LA, Walters TD, et al. Prediction of complicated disease course for children newly diagnosed with Crohn’s disease: a multicentre inception cohort study. Lancet. 2017;389(10080):1710–1718. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Richard AC, Ferdinand JR, Meylan F, Hayes ET, Gabay O, Siegel RM. The TNF-family cytokine TL1A: from lymphocyte costimulator to disease co-conspirator. J Leukoc Biol. 2015;98(3):333–345. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Jacob N, Jacobs JP, Kumagai K, et al. Inflammation-independent TL1A-mediated intestinal fibrosis is dependent on the gut microbiome. Mucosal Immunol. 2018;11(5):1466–1476. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Bamias G, Martin C, 3rd, Marini M, et al. Expression, localization, and functional activity of TL1A, a novel Th1-polarizing cytokine in inflammatory bowel disease. J Immunol. 2003;171(9):4868–4874. [DOI] [PubMed] [Google Scholar]
- 141.Slebioda TJ, Bojarska-Junak A, Stanislawowski M, et al. TL1A as a Potential Local Inducer of IL17A Expression in Colon Mucosa of Inflammatory Bowel Disease Patients. Scand J Immunol. 2015;82(4):352–360. [DOI] [PubMed] [Google Scholar]
- 142.Barrett R, Zhang X, Koon HW, et al. Constitutive TL1A expression under colitogenic conditions modulates the severity and location of gut mucosal inflammation and induces fibrostenosis. Am J Pathol. 2012;180(2):636–649. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Shih DQ, Barrett R, Zhang X, et al. Constitutive TL1A (TNFSF15) expression on lymphoid or myeloid cells leads to mild intestinal inflammation and fibrosis. PLoS One. 2011;6(1):e16090. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Zheng L, Zhang X, Chen J, et al. Sustained Tl1a (Tnfsf15) Expression on Both Lymphoid and Myeloid Cells Leads to Mild Spontaneous Intestinal Inflammation and Fibrosis. Eur J Microbiol Immunol (Bp). 2013;3(1):11–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Li H, Song J, Niu G, et al. TL1A blocking ameliorates intestinal fibrosis in the T cell transfer model of chronic colitis in mice. Pathol Res Pract. 2018;214(2):217–227. [DOI] [PubMed] [Google Scholar]
- 146.Shih DQ, Zheng L, Zhang X, et al. Inhibition of a novel fibrogenic factor Tl1a reverses established colonic fibrosis. Mucosal Immunol. 2014;7(6):1492–1503. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Jacob N, Kumagai K, Abraham JP, et al. Direct signaling of TL1A-DR3 on fibroblasts induces intestinal fibrosis in vivo. Sci Rep. 2020;10(1):18189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Latella G, Viscido A. Controversial Contribution of Th17/IL-17 Toward the Immune Response in Intestinal Fibrosis. Dig Dis Sci. 2020;65(5):1299–1306. [DOI] [PubMed] [Google Scholar]
- 149.Ramani K, Biswas PS. Interleukin-17: Friend or foe in organ fibrosis. Cytokine. 2019;120:282–288. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Xu W, Su L, Qing P, et al. Elevated levels of TL1A are associated with disease activity in patients with systemic sclerosis. Clin Rheumatol. 2017;36(6):1317–1324. [DOI] [PubMed] [Google Scholar]
- 151.Biancheri P, Pender SL, Ammoscato F, et al. The role of interleukin 17 in Crohn’s disease-associated intestinal fibrosis. Fibrogenesis Tissue Repair. 2013;6(1):13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Honzawa Y, Nakase H, Shiokawa M, et al. Involvement of interleukin-17A-induced expression of heat shock protein 47 in intestinal fibrosis in Crohn’s disease. Gut. 2014;63(12):1902–1912. [DOI] [PubMed] [Google Scholar]
- 153.Zhang HJ, Zhang YN, Zhou H, Guan L, Li Y, Sun MJ. IL-17A Promotes Initiation and Development of Intestinal Fibrosis Through EMT. Dig Dis Sci. 2018;63(11):2898–2909. [DOI] [PubMed] [Google Scholar]
- 154.Li J, Liu L, Zhao Q, Chen M. Role of Interleukin-17 in Pathogenesis of Intestinal Fibrosis in Mice. Dig Dis Sci. 2020;65(7):1971–1979. [DOI] [PubMed] [Google Scholar]
- 155.Hueber W, Sands BE, Lewitzky S, et al. Secukinumab, a human anti-IL-17A monoclonal antibody, for moderate to severe Crohn’s disease: unexpected results of a randomised, double-blind placebo-controlled trial. Gut. 2012;61(12):1693–1700. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Li J, Ueno A, Fort Gasia M, et al. Profiles of Lamina Propria T Helper Cell Subsets Discriminate Between Ulcerative Colitis and Crohn’s Disease. Inflamm Bowel Dis. 2016;22(8):1779–1792. [DOI] [PubMed] [Google Scholar]
- 157.Valatas V, Bamias G, Kolios G. Experimental colitis models: Insights into the pathogenesis of inflammatory bowel disease and translational issues. Eur J Pharmacol. 2015;759:253–264. [DOI] [PubMed] [Google Scholar]
- 158.Kugathasan S, Saubermann LJ, Smith L, et al. Mucosal T-cell immunoregulation varies in early and late inflammatory bowel disease. Gut. 2007;56(12):1696–1705. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Postlethwaite AE, Holness MA, Katai H, Raghow R. Human fibroblasts synthesize elevated levels of extracellular matrix proteins in response to interleukin 4. J Clin Invest. 1992;90(4):1479–1485. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Aoudjehane L, Pissaia A, Jr., Scatton O, et al. Interleukin-4 induces the activation and collagen production of cultured human intrahepatic fibroblasts via the STAT-6 pathway. Lab Invest. 2008;88(9):973–985. [DOI] [PubMed] [Google Scholar]
- 161.Niessner M, Volk BA. Altered Th1/Th2 cytokine profiles in the intestinal mucosa of patients with inflammatory bowel disease as assessed by quantitative reversed transcribed polymerase chain reaction (RT-PCR). Clin Exp Immunol. 1995;101(3):428–435. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Boirivant M, Fuss IJ, Chu A, Strober W. Oxazolone colitis: A murine model of T helper cell type 2 colitis treatable with antibodies to interleukin 4. J Exp Med. 1998;188(10):1929–1939. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Fichtner-Feigl S, Strober W, Kawakami K, Puri RK, Kitani A. IL-13 signaling through the IL-13alpha2 receptor is involved in induction of TGF-beta1 production and fibrosis. Nat Med. 2006;12(1):99–106. [DOI] [PubMed] [Google Scholar]
- 164.Fichtner-Feigl S, Young CA, Kitani A, Geissler EK, Schlitt HJ, Strober W. IL-13 signaling via IL-13R alpha2 induces major downstream fibrogenic factors mediating fibrosis in chronic TNBS colitis. Gastroenterology. 2008;135(6):2003–2013, 2013 e2001–2007. [DOI] [PubMed] [Google Scholar]
- 165.Bailey JR, Bland PW, Tarlton JF, et al. IL-13 promotes collagen accumulation in Crohn’s disease fibrosis by down-regulation of fibroblast MMP synthesis: a role for innate lymphoid cells? PLoS One. 2012;7(12):e52332. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Reinisch W, Panes J, Khurana S, et al. Anrukinzumab, an anti-interleukin 13 monoclonal antibody, in active UC: efficacy and safety from a phase IIa randomised multicentre study. Gut. 2015;64(6):894–900. [DOI] [PubMed] [Google Scholar]
- 167.Danese S, Rudzinski J, Brandt W, et al. Tralokinumab for moderate-to-severe UC: a randomised, double-blind, placebo-controlled, phase IIa study. Gut. 2015;64(2):243–249. [DOI] [PubMed] [Google Scholar]
- 168.Guillonneau C, Bezie S, Anegon I. Immunoregulatory properties of the cytokine IL-34. Cell Mol Life Sci. 2017;74(14):2569–2586. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Baghdadi M, Umeyama Y, Hama N, et al. Interleukin-34, a comprehensive review. J Leukoc Biol. 2018;104(5):931–951. [DOI] [PubMed] [Google Scholar]
- 170.Preisser L, Miot C, Le Guillou-Guillemette H, et al. IL-34 and macrophage colony-stimulating factor are overexpressed in hepatitis C virus fibrosis and induce profibrotic macrophages that promote collagen synthesis by hepatic stellate cells. Hepatology. 2014;60(6):1879–1890. [DOI] [PubMed] [Google Scholar]
- 171.Shoji H, Yoshio S, Mano Y, et al. Interleukin-34 as a fibroblast-derived marker of liver fibrosis in patients with non-alcoholic fatty liver disease. Sci Rep. 2016;6:28814. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Wang YQ, Cao WJ, Gao YF, Ye J, Zou GZ. Serum interleukin-34 level can be an indicator of liver fibrosis in patients with chronic hepatitis B virus infection. World J Gastroenterol. 2018;24(12):1312–1320. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Baek JH, Zeng R, Weinmann-Menke J, et al. IL-34 mediates acute kidney injury and worsens subsequent chronic kidney disease. J Clin Invest. 2015;125(8):3198–3214. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Franze E, Dinallo V, Laudisi F, et al. Interleukin-34 Stimulates Gut Fibroblasts to Produce Collagen Synthesis. J Crohns Colitis. 2020;14(10):1436–1445. [DOI] [PubMed] [Google Scholar]
- 175.Zmora N, Suez J, Elinav E. You are what you eat: diet, health and the gut microbiota. Nat Rev Gastroenterol Hepatol. 2019;16(1):35–56. [DOI] [PubMed] [Google Scholar]
- 176.Schirmer M, Garner A, Vlamakis H, Xavier RJ. Microbial genes and pathways in inflammatory bowel disease. Nat Rev Microbiol. 2019;17(8):497–511. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Walsh D, McCarthy J, O’Driscoll C, Melgar S. Pattern recognition receptors--molecular orchestrators of inflammation in inflammatory bowel disease. Cytokine Growth Factor Rev. 2013;24(2):91–104. [DOI] [PubMed] [Google Scholar]
- 178.Rieder F The gut microbiome in intestinal fibrosis: environmental protector or provocateur? Sci Transl Med. 2013;5(190):190ps110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Otte JM, Rosenberg IM, Podolsky DK. Intestinal myofibroblasts in innate immune responses of the intestine. Gastroenterology. 2003;124(7):1866–1878. [DOI] [PubMed] [Google Scholar]
- 180.Bhogal RH, Sutaria R, Afford SC. Bacterial lipopolysaccharide promotes profibrotic activation of intestinal fibroblasts (Br J Surg 2010; 97: 1126–1134). Br J Surg. 2010;97(11):1742; author reply 1742. [DOI] [PubMed] [Google Scholar]
- 181.Jun YK, Kwon SH, Yoon HT, et al. Toll-like receptor 4 regulates intestinal fibrosis via cytokine expression and epithelial-mesenchymal transition. Sci Rep. 2020;10(1):19867. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.Ray S, De Salvo C, Pizarro TT. Central role of IL-17/Th17 immune responses and the gut microbiota in the pathogenesis of intestinal fibrosis. Curr Opin Gastroenterol. 2014;30(6):531–538. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Grassl GA, Valdez Y, Bergstrom KS, Vallance BA, Finlay BB. Chronic enteric salmonella infection in mice leads to severe and persistent intestinal fibrosis. Gastroenterology. 2008;134(3):768–780. [DOI] [PubMed] [Google Scholar]
- 184.Palmela C, Chevarin C, Xu Z, et al. Adherent-invasive Escherichia coli in inflammatory bowel disease. Gut. 2018;67(3):574–587. [DOI] [PubMed] [Google Scholar]
- 185.Darfeuille-Michaud A, Boudeau J, Bulois P, et al. High prevalence of adherent-invasive Escherichia coli associated with ileal mucosa in Crohn’s disease. Gastroenterology. 2004;127(2):412–421. [DOI] [PubMed] [Google Scholar]
- 186.Small CL, Reid-Yu SA, McPhee JB, Coombes BK. Persistent infection with Crohn’s disease-associated adherent-invasive Escherichia coli leads to chronic inflammation and intestinal fibrosis. Nat Commun. 2013;4:1957. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187.Schultz BM, Salazar GA, Paduro CA, et al. Persistent Salmonella enterica serovar Typhimurium Infection Increases the Susceptibility of Mice to Develop Intestinal Inflammation. Front Immunol. 2018;9:1166. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.Zundler S, Becker E, Weidinger C, Siegmund B. Anti-Adhesion Therapies in Inflammatory Bowel Disease-Molecular and Clinical Aspects. Front Immunol. 2017;8:891. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189.Mao R, Kurada S, Gordon IO, et al. The Mesenteric Fat and Intestinal Muscle Interface: Creeping Fat Influencing Stricture Formation in Crohn’s Disease. Inflamm Bowel Dis. 2019;25(3):421–426. [DOI] [PubMed] [Google Scholar]
- 190.Borley NR, Mortensen NJ, Jewell DP, Warren BF. The relationship between inflammatory and serosal connective tissue changes in ileal Crohn’s disease: evidence for a possible causative link. J Pathol. 2000;190(2):196–202. [DOI] [PubMed] [Google Scholar]
- 191.Serena C, Keiran N, Madeira A, et al. Crohn’s Disease Disturbs the Immune Properties of Human Adipose-Derived Stem Cells Related to Inflammasome Activation. Stem Cell Reports. 2017;9(4):1109–1123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Serena C, Millan M, Ejarque M, et al. Adipose stem cells from patients with Crohn’s disease show a distinctive DNA methylation pattern. Clin Epigenetics. 2020;12(1):53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193.Serena C, Queipo-Ortuno M, Millan M, et al. Microbial Signature in Adipose Tissue of Crohn’s Disease Patients. J Clin Med. 2020;9(8). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194.Guedj K, Abitbol Y, Cazals-Hatem D, et al. Adipocytes orchestrate the formation of tertiary lymphoid organs in the creeping fat of Crohn’s disease affected mesentery. J Autoimmun. 2019;103:102281. [DOI] [PubMed] [Google Scholar]
- 195.Bilski J, Mazur-Bialy A, Wojcik D, et al. Role of Obesity, Mesenteric Adipose Tissue, and Adipokines in Inflammatory Bowel Diseases. Biomolecules. 2019;9(12). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196.Rieder F, Doyon G, Ouyang Z, West G, Fiocchi C. 573 Adipocyte and Preadipocyte Derived-Mediators Induce a PRO-Fibrogenic Phenotype in Human Intestinal Mesenchymal Cells -A Novel Link Between Fat and Intestinal Fibrosis. Gastroenterology. 2014;146(5):S-106–S-106. [Google Scholar]
- 197.Mao R, Doyon G, Kurada S, et al. 629 - Creeping-Fat Derived Free Fatty Acids Induce Hyperplasia of Intestinal Muscularis Propria Muscle Cells – A Novel Link Between Fat and Intestinal Stricture Formation in Crohn’s Disease. Gastroenterology. 2018;154(6, Supplement 1):S–131. [Google Scholar]
- 198.Kredel LI, Batra A, Stroh T, et al. Adipokines from local fat cells shape the macrophage compartment of the creeping fat in Crohn’s disease. Gut. 2013;62(6):852–862. [DOI] [PubMed] [Google Scholar]
- 199.Wu J, Lubman DM, Kugathasan S, et al. Serum Protein Biomarkers of Fibrosis Aid in Risk Stratification of Future Stricturing Complications in Pediatric Crohn’s Disease. Am J Gastroenterol. 2019;114(5):777–785. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200.Higgins PD. Measurement of Fibrosis in Crohn’s Disease Strictures with Imaging and Blood Biomarkers to Inform Clinical Decisions. Dig Dis. 2017;35(1–2):32–37. [DOI] [PubMed] [Google Scholar]
- 201.Bettenworth D, Bokemeyer A, Baker M, et al. Assessment of Crohn’s disease-associated small bowel strictures and fibrosis on cross-sectional imaging: a systematic review. Gut. 2019;68(6):1115–1126. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202.Adler J, Punglia DR, Dillman JR, et al. Computed tomography enterography findings correlate with tissue inflammation, not fibrosis in resected small bowel Crohn’s disease. Inflamm Bowel Dis. 2012;18(5):849–856. [DOI] [PubMed] [Google Scholar]
- 203.Rieder F, Feagan BG, Jairath V, group Cs. Editorial: treating strictures in inflammatory bowel disease-authors’ reply. Aliment Pharmacol Ther. 2018;48(11–12):1313–1314. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 204.Wilkens R, Hagemann-Madsen RH, Peters DA, et al. Validity of Contrast-enhanced Ultrasonography and Dynamic Contrast-enhanced MR Enterography in the Assessment of Transmural Activity and Fibrosis in Crohn’s Disease. J Crohns Colitis. 2018;12(1):48–56. [DOI] [PubMed] [Google Scholar]
- 205.Chiorean MV, Sandrasegaran K, Saxena R, Maglinte DD, Nakeeb A, Johnson CS. Correlation of CT enteroclysis with surgical pathology in Crohn’s disease. Am J Gastroenterol. 2007;102(11):2541–2550. [DOI] [PubMed] [Google Scholar]
- 206.Jensen MR, Friberg L, Karlsmark T, Bulow J. (18)F-FDG PET/CT in a rare case of Stewart-Treves syndrome: future implications and diagnostic considerations. Lymphat Res Biol. 2011;9(1):61–64. [DOI] [PubMed] [Google Scholar]
- 207.Soyer P, Boudiaf M, Dray X, et al. Crohn’s disease: multi-detector row CT-enteroclysis appearance of the appendix. Abdom Imaging. 2010;35(6):654–660. [DOI] [PubMed] [Google Scholar]
- 208.Wagner M, Ko HM, Chatterji M, et al. Magnetic Resonance Imaging Predicts Histopathological Composition of Ileal Crohn’s Disease. J Crohns Colitis. 2018;12(6):718–729. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 209.Punwani S, Rodriguez-Justo M, Bainbridge A, et al. Mural inflammation in Crohn disease: location-matched histologic validation of MR imaging features. Radiology. 2009;252(3):712–720. [DOI] [PubMed] [Google Scholar]
- 210.Steward MJ, Punwani S, Proctor I, et al. Non-perforating small bowel Crohn’s disease assessed by MRI enterography: derivation and histopathological validation of an MR-based activity index. Eur J Radiol. 2012;81(9):2080–2088. [DOI] [PubMed] [Google Scholar]
- 211.Catalano OA, Gee MS, Nicolai E, et al. Evaluation of Quantitative PET/MR Enterography Biomarkers for Discrimination of Inflammatory Strictures from Fibrotic Strictures in Crohn Disease. Radiology. 2016;278(3):792–800. [DOI] [PubMed] [Google Scholar]
- 212.Guyatt GH, Kirshner B, Jaeschke R. Measuring health status: what are the necessary measurement properties? J Clin Epidemiol. 1992;45(12):1341–1345. [DOI] [PubMed] [Google Scholar]
- 213.Gordon IO, Bettenworth D, Bokemeyer A, et al. Histopathology Scoring Systems of Stenosis Associated With Small Bowel Crohn’s Disease: A Systematic Review. Gastroenterology. 2020;158(1):137–150 e131. [DOI] [PMC free article] [PubMed] [Google Scholar]