Background: Crohn’s disease (CD) is a condition on the spectrum of inflammatory bowel disease that affects up to 20 people per 100,000 in the US annually, and with incidence increasing. One of the most significant sources of morbidity in CD is the formation of strictures, with resultant intestinal blockage a common indication for hospitalization and surgical intervention in these patients. The pathophysiology of stricture formation is not fully understood. However, the fibroplasia that leads to fibrostenotic stricture formation may have shared pathophysiology with IgG4-related fibrosis. Summary: Initial intestinal inflammation recruits innate immune cells, such as neutrophils, that secrete IL-1β and IL-23, which induces a type 17 CD4+ T-helper T-cell (Th17)-mediated adaptive immune response. These CD4+ Th17 T cells also contribute to inflammation by secreting proinflammatory cytokines such as IL-17 and IL-21. IL-21 recruits and stimulates CD4+ T follicular helper (Tfh) cells, which secrete more IL-21. This causes ectopic germinal center formation, recruiting and stimulating naïve B cells. The IL-17 and IL-21 produced by Th17 cells and Tfh cells also induce IgG4 plasmablast differentiation. Finally, these IgG4-producing plasmablasts secrete platelet-derived growth factor (PDGF), which activates local PDGF-receptor expressing fibroblasts and myofibroblasts, resulting in uncontrolled fibroplasia.

Key Messages

  • Strictures represent a significant source of morbidity in Crohn’s disease (CD).

  • The pathophysiology of CD and IgG4-related diseases share a hyperactive type 17 CD4+ T-helper T-cell response, IL-21 production and converge on sustained, aberrant plasmablast activity.

  • IgG4-producing plasmablasts secrete platelet-derived growth factor, which may cause intestinal fibroplasia and eventual stricture formation.

Crohn’s disease (CD) is a condition on the spectrum of inflammatory bowel disease (IBD) that affects up to 20 people per 100,000 in the US annually, and with incidence increasing. Although having a relapsing and remitting course, CD is a chronic condition with significant associated morbidity and high cost-of-care. One of the most significant sources of morbidity in CD is the formation of strictures, with resultant intestinal blockage a common indication for hospitalization and surgical intervention in these patients. The pathophysiology of stricture formation is not fully understood. However, the fibroplasia that leads to fibrostenotic stricture formation may have shared pathophysiology with IgG4-related fibrosis. The goal of this review is to explore the shared pathophysiology and immunologic mediators of both fibrostenotic stricture formation in CD and IgG4-related diseases and discuss implications on patient course and potential modes of immunotherapy.

CD is a condition on the spectrum of IBD that is characterized by transmural bowel inflammation that may occur in all segments of the gastrointestinal (GI) tract [1-3]. CD affects up to 20 people per 100,000 in the US annually, and the prevalence is increasing [4-6]. CD is a chronic condition that usually follows a relapsing and remitting course. However, the disease is ultimately progressive and is thus associated with significant morbidity and high cost-of-care [7, 8]. Furthermore, all-cause mortality is elevated in CD patients, underscoring the importance of further research to improve patient outcomes [9, 10].

The presentation of CD is varied but usually affects patients between the second and fourth decades, although recent studies have suggested a second age peak of presentation in older patients [11, 12]. The most common presenting symptoms of CD include fatigue, abdominal pain, diarrhea, and bloody bowel movements [7, 8, 11]. Although nonspecific, these symptoms are generally what cause patients to present to medical attention. The diagnosis of CD is made through a combination of endoscopic and imaging findings usually showing noncontiguous lesions throughout the GI tract, as well as biopsy and histopathologic evaluation with granulomas and transmural inflammation supportive of the diagnosis of CD [5, 13].

Symptoms may progress and result in complications such as GI abscesses, fistulas, and strictures [7, 8]. The underlying pathophysiology driving disease progression to these serious complications is likely multifactorial but is driven by mucosal immune dysregulation and inflammation within the bowel. Classically, CD is thought to be driven by Th1-mediated inflammation, with interferon gamma (IFN-γ), tumor necrosis factor (TNF)-alpha (TNF-α) release sustaining inflammation and resulting in granuloma formation [14-16]. However, more recent discoveries have implicated aberrant innate immune responses to the gut microbiome as well as involvement of the type 17 CD4+ T-helper T-cell (Th17)-IL-23 axis as contributing to the pathogenesis of CD [3, 14, 17-19].

Although sharing fundamental immune mechanisms, three distinct phenotypes of CD have been described: stricturing CD, penetrating CD, and non-stricturing non-penetrating CD [20]. These distinctions describe the primary course of disease; however, there is overlap between types [21, 22].

Strictures are a significant source of morbidity, with up to 50% of all CD patients eventually developing strictures that commonly present as obstructive symptoms [8, 21-23]. Several predictors of stricturing CD have been posited, with age of onset under 40 years, perianal involvement at diagnosis, small bowel disease location, and needing steroids during the index CD flare being associated with stricture formation [23]. Furthermore, other complications such as fistula formation have been shown to predict strictures in CD, as well, suggesting advanced disease yields progressive complications [23, 24]. Although no precise definition exists, strictures are frequently diagnosed via radiographic imaging showing narrowing of the intestinal lumen [25-27]. Bowel obstruction in these patients often results in hospitalization, with a large proportion requiring surgical intervention [28, 29].

Previously, strictures have been thought to result from bowel inflammation leading to dysregulated wound healing and resultant scar and fibrosis formation [3, 28]. However, more recent research has identified two processes by which strictures may form; both wound healing secondary to inflammation and independent fibroplasia may lead to stricture formation [29, 30]. As such, strictures can be subclassified as inflammatory strictures or fibrostenotic strictures [30, 31]. Although the clinical presentations of these types of strictures are similar, the difference in underlying pathophysiology necessitates different treatment approaches that, importantly, carry different success rates.

Inflammatory strictures that are a result of dysregulated wound healing secondary to bowel inflammation and injury respond well to existing anti-inflammatory therapy [32, 33]. However, anti-inflammatory therapy in CD is ineffective for reducing intestinal fibrosis and is thus an ineffective modality for fibrostenotic strictures. Anti-inflammatory therapy has been shown to not reduce the need for surgery in CD patients with strictures, with fibrosis continuing even after disease remission [8, 23, 33, 34]. As such, the mainstay of treatment for fibrostenosing strictures is surgical intervention, with endoscopic balloon therapy, stricturoplasties, or resection being employed depending on the severity of symptoms and the degree of stenosis of the intestinal lumen [35-37]. With no existing effective medical therapy for fibrostenosing strictures in CD, further elucidation of the underlying etiology and pathogenesis of intestinal fibroplasia is paramount to developing effective medical treatments for this complication. This would reduce the burden of current surgical treatments placed on patients and allow for earlier initiation of treatment.

The current understanding of the etiology of stricture formation is multifactorial, with a role for both inflammation and fibroplasia that eventually contributes to the narrowing and stenosis of intestines. As mentioned, the inflammatory component is a result of immune dysregulation within the mucosal epithelium of the intestine. It is thought that inflammation in CD begins with an aberrant innate immune response, with a lack of tolerance to native intestinal microbial flora posited as the inciting spark for this inflammatory response [3, 4, 18, 38-40]. This results in innate immune cells, such as neutrophils, dendritic cells, and macrophages, localizing to the lamina propria of the intestine and secreting proinflammatory cytokines such as TNF-α, IFN-γ, and interleukins (IL) 1β, 6, 12, and 23. Innate immune cells within the intestinal lumen also produce these cytokines [1, 3, 18, 38-40] (Fig. 1). IFN-γ and IL-12 induce a Th1-mediated adaptive immune response, with Th1 helper T cells in turn producing more inflammatory cytokines such as TNF-α and IFN-γ as well as IL-2, which sustains a continued Th1 response [1, 3, 18, 38-40]. IL-1β and IL-23 induce a type 17 CD4+ Th17-mediated adaptive immune response. These CD4+ Th17 T cells also contribute to inflammation by secreting proinflammatory cytokines such as IL-17 and IL-21 [14, 18, 41]. In both of these Th1 and Th17 pathways, cytokines produced sustain further differentiation of CD4+ T cells into Th1 and Th17 cells, respectively (Fig. 1).

Fig. 1.

Initial intestinal inflammation recruits innate immune cells, such as neutrophils, that release pro-inflammatory cytokines such as IL-1β and IL-23. These cytokines induce naïve CD4+ T lymphocytes to differentiate into Th17 helper cells. These cells then release cytokines such as IL-17 and IL-21 that in turn induce the differentiation of other naïve CD4+ T lymphocytes into Th17 helper cells, contributing to a self-sustaining Th17 response. The IL-21 released by these cells also recruits Tfh cells, which in turn release more IL-21. IL-17 and IL-21 also induce the formation of ectopic germinal centers, and then further induce naïve B cells within these centers to differentiate into IgG4 producing plasmablasts. Finally, these plasmablasts secrete pro-fibrotic factors such as PDGF that contribute to intestinal fibroplasia.

Fig. 1.

Initial intestinal inflammation recruits innate immune cells, such as neutrophils, that release pro-inflammatory cytokines such as IL-1β and IL-23. These cytokines induce naïve CD4+ T lymphocytes to differentiate into Th17 helper cells. These cells then release cytokines such as IL-17 and IL-21 that in turn induce the differentiation of other naïve CD4+ T lymphocytes into Th17 helper cells, contributing to a self-sustaining Th17 response. The IL-21 released by these cells also recruits Tfh cells, which in turn release more IL-21. IL-17 and IL-21 also induce the formation of ectopic germinal centers, and then further induce naïve B cells within these centers to differentiate into IgG4 producing plasmablasts. Finally, these plasmablasts secrete pro-fibrotic factors such as PDGF that contribute to intestinal fibroplasia.

Close modal

This inflammation results in epithelial and endothelial injury, leading to both anti-inflammatory regulatory responses as well as dysregulated wound healing. In response to proinflammatory cytokines such as TNF-α, IL-6, IFN-γ, and TGF-β, anti-inflammatory cytokines are produced by local immune cells as well as epithelial cells, adipocytes, and fibroblasts [29, 42-44]. These anti-inflammatory cytokines include transforming growth factor β (TGF-β), which is thought to be a main effector of further anti-inflammatory responses and fibroplasia. TGF-β promotes the differentiation of naïve helper T cells into regulatory T cells (Tregs), which in turn secrete anti-inflammatory cytokines [29, 40, 42-44]. However, this response may be tempered in the presence of IL-1β, IL-6, and IL-21, which, along with TGF-β, promote differentiation into Th17 cells [41].

TGF-β also causes the activation and proliferation of local mesenchymal effectors of fibrosis, such as fibroblasts and myofibroblasts. Similarly, it causes recruitment of extraintestinal fibroblasts and myofibroblasts [29, 42-44]. These mesenchymal cells and recruited cells produce collagen and other elements of the extracellular matrix (ECM) [29, 42-45]. Furthermore, TGF-β has been shown to cause transformation of local epithelial and endothelial cells into mesenchymal cells, further expanding the available fibroblasts and myofibroblasts to produce ECM proteins [43]. In physiologic wound healing, this fibroplasia and the production of ECM proteins would be downregulated, while in the context of constant inflammation in CD, these processes become disordered and are continually activated.

What is less clear is the underlying mechanisms that cause progression of fibrosis even in periods of disease treatment and remission of inflammation. One proposed explanation is that the ECM produced in initial stages of fibrosis is a reservoir for further fibrogenic cytokines, such as TGF-β, which may be released even after cessation of inflammation and injury [42, 43]. This release of latent fibrogenic cytokines may contribute to increasing stiffness of the ECM and further crosslinking of existing ECM proteins [46, 47]. This increasing stiffness has been shown to independently promote fibrogenesis even without further fibrogenic stimuli [48-50].

A second proposed explanation is that initial injury and destruction of the epithelial intestinal barrier exposes mesenchymal cells to gut microbiota. These cells are not equipped with tolerance mechanisms usually found in physiologic epithelial cells, and thus are continually activated even in the absence of Th1 and Th17 mediated inflammation. As such, these mesenchymal cells may produce further ECM proteins and contribute to fibrogenesis [51, 52]. However, other reasons for the continually increasing burden of fibrosis over the course of even well-controlled CD are unclear.

The involvement of localized IgG4-related fibroplasia may be a possible explanation for continued fibrosis in CD. IgG4-related disease is characterized by systemic fibrosis that may affect a variety of locations, with the most commonly described being autoimmune pancreatitis, cholangitis, aortitis, retroperitoneal fibrosis, and salivary gland fibrosis [53]. Although the clinical manifestations and histopathologic features of this disease are well characterized, its pathophysiology and the underlying etiology of uncontrolled fibroplasia that characterizes IgG4-related disease are not fully elucidated [53-55].

The current understanding of the pathophysiology of these IgG4-related diseases (IgG4RD) is that T-cell-mediated inflammation results in both the aberrant and extensive production of IgG4 antibodies and in systemic fibroplasia [53-55]. It has been suggested that the IgG4 antibodies have anti-inflammatory properties, suggesting that they are not pathogenic, but possibly reactive to an upstream process [56]. Characterizing the inflammation that is the nidus for this disease has proven difficult, although several mechanisms have been proposed. The first is an autoimmune response to self-antigens, although this hypothesis has not been widely confirmed across the spectrum of IgG4RD [57-59].

Although the initiation of the systemic inflammation has not been definitively found, much progress understanding the effectors of this inflammation has been made. One cell type thought to be involved with the pathogenesis of IgG4RD is the CD4+ cytotoxic T cell. These cells are able to secrete granzyme B and perforin, which allows for cytotoxic activity while interacting with major histocompatibility II-expressing cells [60]. Elevations in these cells, both in lesional tissue and in peripheral blood, are found in patients with IgG4RD and are thought to contribute to disease pathogenesis by secreting IFNγ, IL-1β, and TGF-β, all of which are known to activate fibroblasts [61]. Notably, populations of these cells have been shown to decrease in patients successfully treated with glucocorticoids, suggesting a role for this cell subtype in disease progression [62-65].

A second cell type central to the pathogenesis of IgG4RD is the CD4+ follicular helper T cell (Tfh). These cells have also been shown to be elevated in both the peripheral blood and within involved tissue in patients with IgG4RD [66, 67]. Furthermore, these cells have been shown to have a reciprocal relationship with CD4+ cytotoxic T cells. These cells both differentiate from naïve CD4+ lymphocytes, but the differentiation of each cell type is inversely proportional and dependent on local immune milieu [68]. Tfh cells have been shown to cause the formation of pseudo germinal centers within affected tissue, and their presence correlates with plasmablast class switching, proliferation, and production of IgG4 antibodies [62, 69-71].

Finally, IgG4-secreting plasmablasts are another effector cell of interest in the pathogenesis of IgG4RD. These cells may act as antigen-presenting cells, sustaining the above described population of CD4+ cytotoxic T cells and thus contributing to sustained inflammation and fibrogenesis [65, 72]. Further, these cells may directly contribute to the fibrosis seen in IgG4RD. Plasmablasts may secrete platelet-derived growth factor (PDGF), which is known to activate fibroblasts and thus significantly contribute to fibrosis [73-78] The central role of these plasmablasts in IgG4RD is emphasized by the efficacy of rituxumab in disease treatment, and disease flares coinciding with an increase in IgG4 producing plasmablasts [72, 73, 79-84].

Although the understanding of systemic IgG4 disease is improving, there is little reported on the influence of IgG4 mechanisms of disease on localized fibroplasia. It is possible that some or all of these mechanisms may be involved in localized fibroplasia, and further research on this topic is warranted.

The involvement of IgG4 mechanisms within the fibrosis seen in CD is currently unclear. Although there has been evidence to suggest mucosal infiltration of IgG4 plasma cells in patients with ulcerative colitis, there is a dearth of research on a similar process in CD [13, 74-77]. Further, there is little investigation into the contribution of IgG4 mechanisms in stricturing CD. However, there are several plausible shared immunologic mechanisms that warrant future study in the context of stricture formation and fibrosis.

The first shared mechanism of interest is the interplay between initial inflammation, Th17 response, and follicular T-cell recruitment. As mentioned, initial epithelial injury and innate immune system response within intestinal mucosa results in the release of pro-inflammatory cytokines IL-1β, IL-6, and IL-21. These cytokines, along with TGF-B, released as part of the initial anti-inflammatory response, drive naïve CD4+ T cells to differentiate into Th17 effector cells [41]. IL-21 is particularly of interest, as it is a strong inducer of this Th17 response. Th17 cells further produce IL-21, resulting in a self-sustaining immune response [78, 85-88]. The role of IL-21 in CD is established, with evidence showing IL-21 is elevated in those with CD, specifically within samples of CD lesions, and that it contributes to a sustained, aberrant inflammatory immune response in intestinal mucosa [89-92].

This Th17/IL-21 interplay is also relevant for the recruitment of follicular T cells and the formation of germinal centers, both central effectors of IgG4RD. First, IL-21 has been shown to stimulate Tfh cells, which in turn produce more IL-21 [92, 93]. This, again, further stimulates Th17 differentiation and thus IL-21 release by Th17 cells. Tfh cells and their secretion of IL-21 also promote the formation of germinal centers [94-100]. Il-17 and the Th17 mediated immune response has also been shown to promote germinal center formation, complimenting the IL-21 mediated response [101-103]. Finally, and crucially, cytokines secreted by Tfh cells, namely, IL-21, also promotes differentiation of naïve B cells within germinal centers into IgG4 producing plasmablasts. This association has been strengthened by the localization of IL-21-secreting Tfh cells within germinal centers in IgG4RD [104-107]. Similarly, hyperactive B cells that preferentially produce IgG class antibodies have been described within granulomas in CD, lending credence to plasmablasts as a final, shared result of the Th17/IL-21 axis [108, 109] (Fig. 1).

Both IgG4RD and CD have shared mechanisms that converge on the formation of plasmablast-rich germinal centers. This convergence may finally relate to fibrosis through the plasmablast-mediated release of PDGF as described above, with PDGF causing fibroblast activation and proliferation, leading to fibrosis [73, 79-84]. Fibrosis has been reported within and surrounding germinal centers containing IgG4 plasma cells in both IgG4RD and other inflammatory conditions, potentially providing evidence of plasma cell-induced fibroplasia [110-114]. PDGF receptor-positive cells have also been described within the GI tract and PDGF has also been localized to active inflammation and active fibrosis within CD, possibly explaining the widespread fibrosis seen within some CD [115-117] (Fig. 1).

These mechanisms provide a plausible role for localized IgG4-related fibrosis within CD, and may contribute to fibrostenotic stricture formation. Furthermore, these mechanisms are not dependent on targets of current CD medical management [3]. This may explain the progression of fibrosis even in well-controlled CD in which inflammation has abated. Although further research is needed to clarify this potential relationship, localized IgG4 involvement and fibrosis as a mechanism for fibrostenotic stricture formation in CD is a promising area of future investigation.

Additionally, as IgG4 plasmablasts have been identified in the intestinal epithelium in ulcerative colitis, it is plausible that they may be responsible for colorectal strictures in UC [13, 74-76]. Although less significant in UC, potentially due to the larger caliber of the colon compared to the small intestine, IgG4 fibroplasia may be at play in UC, as well [118, 119]. Future research is warranted to clarify the significance of IgG4 plasmablasts in UC.

Along with the above shared pathophysiology, auxiliary immunologic mechanisms likely also contribute to the formation of IgG4-secreting plasmablasts and thus the fibrosis seen in CD. The first of these mechanisms worth noting is the significance of IL-10 as a contributor to not only fibrosis but also IgG4 plasmablast differentiation. IL-10 may be secreted from several involved cellular effectors within CD. The first worth noting is Tregs, which are promoted by the secretion of TGF-β [29, 40, 42-44]. Although, as mentioned above, differentiation of naive T cells into Tregs is likely tempered by the presence of IL-1β, IL-6, and IL-21, which shunt this differentiation towards Th17 cells rather than Tregs [41], some Tregs are likely formed. These Tregs then secrete IL-10, a strong inducer of IgG4 plasmablast differentiation, and TGF-β, which promotes local fibrosis. Tregs have been linked to IgG4RD, with underlying pathophysiology posited to be due to this IL-10 and TGF-β secretion [120-127].

Another immunologic pathway that may contribute to IgG4 mediated fibrosis is the release and downstream effects of IL-33 within the gut mucosa. IL-33, or alarmin, is found within epithelial barriers, and, when epithelium is disrupted, IL-33 is released [128]. IL-33 has been shown to be overexpressed in patients with IBD, likely as a result of continuous inflammation causing injury to gut epithelium [129-131]. IL-33 is widely known as a strong inducer of a Th2 response; however, it is critical in the activation of macrophages implicated in wound healing [132, 133]. These macrophages, known as M2 macrophages, arise in the setting of injury and secrete IL-10 and TGF-β, further contributing to Treg differentiation, IgG4 plasmablast differentiation, and fibrosis [134-136]. IL-33 has also been shown to promote Treg differentiation, again contributing to the above mechanism [137]. Although a more recent development, IL-33 has been linked to IgG4RD, strengthening the possible relationship between Il-33 release from injured gut epithelium and IgG4 mediated fibrosis [138, 139].

These mechanisms remain promising to explain how fibrosis and resultant stricture formation may be due to dysregulated wound healing. These processes are also shared by IgG4RD, lending credence to their explanation of IgG4 fibrosis in CD. However, these mechanisms would not explain why fibrosis continues even in the absence of active inflammation, which is central to the significance and morbidity of CD strictures. As such, although they may accelerate or worsen fibroplasia during periods of active disease, these mechanisms are unlikely to be primary immunologic effectors of fibrous stricture formation. Furthermore, these mechanisms are not specific to CD, and rather represent general responses to inflammation. Thus, they may be less likely culprits for the significant fibrostenotic stricture formation seen specifically in CD. For example, gut inflammation and mucosal damage seen in ulcerative colitis would yield similar anti-inflammatory IL-10 secretion and release of IL-33; however, strictures are much rarer and more benign [140]. However, their role may be as a nidus for IgG4 differentiation and fibroplasia within CD. Future research investigating the interplay of these mechanisms and fibrostenotic stricture formation in CD is warranted.

Although confirmatory research must be conducted prior to targeting Th17, Tfh, and IgG4 plasmablasts and their effector cytokines in treatments of fibrosis in CD, several salient targets for immunotherapy emerge. First, treatments for IgG4RD, such as rituximab, may be of use. Rituximab targets CD20 on B cells and prevents their differentiation into plasmablasts, thereby preventing PDGF-mediated fibroplasia as described above. Rituximab has proven effective and safe in small trials for IgG4RD, and this may be repurposed for localized IgG4 fibrosis within stricturing CD [73, 141, 142]. Similarly, next-generation biologics that target CD20, such as ocrelizumab, obinutuzumab, and veltuzumab, may be similarly effective [143]. However, rituximab has been implicated in GI complications and reports of rituximab-induced colitis [144-146]. Further, rituximab may target follicular B cells expressing CD20 and prevent their differentiation into plasmablasts, but would not target chronically active plasmablasts already formed prior to initiation of treatment [147].

Chronically active plasmablasts express CD38, and so the anti-CD38 biologic daratumumab may be a more effective treatment target. In a study of its efficacy in treating patients with multiple myeloma, it has been shown to decrease levels of plasmablasts in healthy controls [148]. Additionally, leveraging the overproduction of IgG4 antibodies in plasmablasts within fibrostenotic strictures by utilizing a proteasome inhibitor such as bortezomib may also be promising. One case of IgG4RD has been shown to be successfully treated with bortezomib [149]. Both daratumumab and bortezomib have shown efficacy in the treatment of multiple myeloma, a plasma cell-driven disease, and so may be effective in treating plasmablast-driven intestinal fibroplasia, as well. Finally, targeting PDGF that is secreted from plasmablasts itself as a final pro-fibrotic effector may be possible. There is little evidence for PDGF as a therapeutic target, and there is no current clinical use. However, preclinical studies have shown a reduction in fibrosis [150, 151].

Blockade of the cytokines involved in the shared pathophysiology of CD and IgG4-related fibrosis also could be an effective treatment for fibrostenotic strictures. Further, anti-IL-21 or anti-17 directed medication may be efficacious in controlling fibrosis. IL-17 blockade is effective and has a favorable safety profile in the treatment of psoriasis; however it has been paradoxically linked to worsening CD flares, and thus may not be the optimal target in this case, although the mechanisms underlying this association are not clear [152-154]. IL-21 seems a more promising target, as anti-IL-21 drugs have been shown to reduce inflammation and neutrophil infiltration in CD. Thus, it may reduce CD inflammation while also targeting a key effector in sustaining IgG4-producing plasma cells [155-157].

The shared pathophysiology of IgG4-related disease and CD provide a plausible mechanism by which IgG4-related fibrosis significantly contributes to fibroplasia and resultant stricture formation. As such, what is currently a progressive and significant source of morbidity in CD may be treatable with immunotherapy directed at the mediators of intestinal fibroplasia. However, although promising, further research into IgG4-related fibrosis and the formation of strictures in CD is imperative to fully elucidate its pathophysiology and facilitate the development of effective treatments.

The authors have no conflicts of interest to declare.

No funding was used for this article.

All authors were involved in the conceptual design of this review. G.-Y.Y. as correspondence author oversaw and supervised entire project, conceptual design, and overall writing and revision. O.B. wrote the initial draft of the review. O.B., D.J.E., and S.E.W. made the initial figure for the review. All authors revised the manuscript and revised the figure. All authors reviewed and approved the final draft of the article.

1.
Gajendran
M
,
Loganathan
P
,
Catinella
AP
,
Hashash
JG
.
A comprehensive review and update on Crohn’s disease
.
Dis Mon
.
2018
;
64
(
2
):
20
57
. .
2.
Kalla
R
,
Ventham
NT
,
Satsangi
J
,
Arnott
ID
.
Crohn’s disease
.
BMJ
.
2014
;
349
:
g6670
. .
3.
Abraham
C
,
Cho
JH
.
Inflammatory bowel disease
.
N Engl J Med
.
2009
;
361
(
21
):
2066
78
. .
4.
Torres
J
,
Mehandru
S
,
Colombel
JF
,
Peyrin-Biroulet
L
.
Crohn’s disease
.
Lancet
.
2017
;
389
(
10080
):
1741
55
. .
5.
Feuerstein
JD
,
Cheifetz
AS
.
Crohn disease: epidemiology, diagnosis, and management
.
Mayo Clin Proc
.
2017
;
92
(
7
):
1088
103
. .
6.
Molodecky
NA
,
Soon
IS
,
Rabi
DM
,
Ghali
WA
,
Ferris
M
,
Chernoff
G
,
Increasing incidence and prevalence of the inflammatory bowel diseases with time, based on systematic review
.
Gastroenterology
.
2012
;
142
(
1
):
46
54.e42
; quiz e30.
7.
Aniwan
S
,
Park
SH
,
Loftus
EV
.
Epidemiology, Natural history, and risk stratification of Crohn’s disease
.
Gastroenterol Clin North Am
.
2017
;
46
(
3
):
463
80
. .
8.
Freeman
HJ
.
Natural history and long-term clinical course of Crohn’s disease
.
World J Gastroenterol
.
2014
;
20
(
1
):
31
6
. .
9.
Wolters
FL
,
Russel
MG
,
Sijbrandij
J
,
Schouten
LJ
,
Odes
S
,
Riis
L
,
Crohn’s disease: increased mortality 10 years after diagnosis in a Europe-wide population based cohort
.
Gut
.
2006
;
55
(
4
):
510
8
.
10.
Loftus
EV
 Jr
.
A matter of life or death: mortality in Crohn’s disease
.
Inflamm Bowel Dis
.
2002
;
8
(
6
):
428
9
. .
11.
Perler
BK
,
Ungaro
R
,
Baird
G
,
Mallette
M
,
Bright
R
,
Shah
S
,
Presenting symptoms in inflammatory bowel disease: descriptive analysis of a community-based inception cohort
.
BMC Gastroenterol
.
2019
;
19
(
1
):
47
.
12.
Hou
JK
,
Kramer
JR
,
Richardson
P
,
Mei
M
,
El-Serag
HB
.
The incidence and prevalence of inflammatory bowel disease among US veterans: a national cohort study
.
Inflamm Bowel Dis
.
2013
;
19
(
5
):
1059
64
. .
13.
Magro
F
,
Langner
C
,
Driessen
A
,
Ensari
A
,
Geboes
K
,
Mantzaris
GJ
,
European consensus on the histopathology of inflammatory bowel disease
.
J Crohns Colitis
.
2013
;
7
(
10
):
827
51
.
14.
Brand
S
.
Crohn’s disease: Th1, Th17 or both? The change of a paradigm: new immunological and genetic insights implicate Th17 cells in the pathogenesis of Crohn’s disease
.
Gut
.
2009
;
58
(
8
):
1152
67
. .
15.
Kakazu
T
,
Hara
J
,
Matsumoto
T
,
Nakamura
S
,
Oshitani
N
,
Arakawa
T
,
Type 1 T-helper cell predominance in granulomas of Crohn’s disease
.
Am J Gastroenterol
.
1999
;
94
(
8
):
2149
55
.
16.
Molnár
T
,
Tiszlavicz
L
,
Gyulai
C
,
Nagy
F
,
Lonovics
J
.
Clinical significance of granuloma in Crohn’s disease
.
World J Gastroenterol
.
2005
;
11
(
20
):
3118
21
. .
17.
Nemeth
ZH
,
Bogdanovski
DA
,
Barratt-Stopper
P
,
Paglinco
SR
,
Antonioli
L
,
Rolandelli
RH
.
Crohn’s disease and ulcerative colitis show unique cytokine profiles
.
Cureus
.
2017
;
9
(
4
):
e1177
. .
18.
Lee
SH
,
Kwon
JE
,
Cho
ML
.
Immunological pathogenesis of inflammatory bowel disease
.
Intest Res
.
2018
;
16
(
1
):
26
42
. .
19.
Imam
T
,
Park
S
,
Kaplan
MH
,
Olson
MR
.
Effector T helper cell subsets in inflammatory bowel diseases
.
Front Immunol
.
2018
;
9
:
1212
. .
20.
Gasche
C
,
Scholmerich
J
,
Brynskov
J
,
D’Haens
G
,
Hanauer
SB
,
Irvine
EJ
,
A simple classification of Crohn’s disease: report of the working party for the world congresses of gastroenterology, Vienna 1998
.
Inflamm Bowel Dis
.
2000
;
6
(
1
):
8
15
.
21.
Freeman
HJ
.
Application of the Vienna classification for Crohn’s disease to a single clinician database of 877 patients
.
Can J Gastroenterol
.
2001
;
15
(
2
):
89
93
. .
22.
Freeman
HJ
.
Application of the Montreal classification for Crohn’s disease to a single clinician database of 1015 patients
.
Can J Gastroenterol
.
2007
;
21
(
6
):
363
6
. .
23.
Rieder
F
,
Zimmermann
EM
,
Remzi
FH
,
Sandborn
WJ
.
Crohn’s disease complicated by strictures: a systematic review
.
Gut
.
2013
;
62
(
7
):
1072
84
. .
24.
Jürgens
M
,
Brand
S
,
Laubender
RP
,
Seiderer
J
,
Glas
J
,
Wetzke
M
,
The presence of fistulas and NOD2 homozygosity strongly predict intestinal stenosis in Crohn’s disease independent of the IL23R genotype
.
J Gastroenterol
.
2010
;
45
(
7
):
721
31
.
25.
Gasche
C
,
Moser
G
,
Turetschek
K
,
Schober
E
,
Moeschl
P
,
Oberhuber
G
.
Transabdominal bowel sonography for the detection of intestinal complications in Crohn’s disease
.
Gut
.
1999
;
44
(
1
):
112
7
. .
26.
Fiorino
G
,
Bonifacio
C
,
Peyrin-Biroulet
L
,
Minuti
F
,
Repici
A
,
Spinelli
A
,
Prospective comparison of computed tomography enterography and magnetic resonance enterography for assessment of disease activity and complications in ileocolonic Crohn’s disease
.
Inflamm Bowel Dis
.
2011
;
17
(
5
):
1073
80
.
27.
Voderholzer
WA
,
Beinhoelzl
J
,
Rogalla
P
,
Murrer
S
,
Schachschal
G
,
Lochs
H
,
Small bowel involvement in Crohn’s disease: a prospective comparison of wireless capsule endoscopy and computed tomography enteroclysis
.
Gut
.
2005
;
54
(
3
):
369
73
.
28.
Chan
WPW
,
Mourad
F
,
Leong
RW
.
Crohn’s disease associated strictures
.
J Gastroenterol Hepatol
.
2018
;
33
(
5
):
998
1008
. .
29.
Crespi
M
,
Dulbecco
P
,
De Ceglie
A
,
Conio
M
.
Strictures in Crohn’s disease: from pathophysiology to treatment
.
Dig Dis Sci
.
2020
;
65
(
7
):
1904
16
. .
30.
Bettenworth
D
,
Nowacki
TM
,
Cordes
F
,
Buerke
B
,
Lenze
F
.
Assessment of stricturing Crohn’s disease: current clinical practice and future avenues
.
World J Gastroenterol
.
2016
;
22
(
3
):
1008
16
. .
31.
Lenze
F
,
Wessling
J
,
Bremer
J
,
Ullerich
H
,
Spieker
T
,
Weckesser
M
,
Detection and differentiation of inflammatory versus fibromatous Crohn’s disease strictures: prospective comparison of 18F-FDG-PET/CT, MR-enteroclysis, and transabdominal ultrasound versus endoscopic/histologic evaluation
.
Inflamm Bowel Dis
.
2012
;
18
(
12
):
2252
60
.
32.
Sorrentino
D
,
Avellini
C
,
Beltrami
CA
,
Pasqual
E
,
Zearo
E
.
Selective effect of infliximab on the inflammatory component of a colonic stricture in Crohn’s disease
.
Int J Colorectal Dis
.
2006
;
21
(
3
):
276
81
. .
33.
Holtmann
MH
,
Neurath
MF
.
Anti-TNF strategies in stenosing and fistulizing Crohn’s disease
.
Int J Colorectal Dis
.
2005
;
20
(
1
):
1
8
. .
34.
Cosnes
J
,
Nion-Larmurier
I
,
Beaugerie
L
,
Afchain
P
,
Tiret
E
,
Gendre
JP
.
Impact of the increasing use of immunosuppressants in Crohn’s disease on the need for intestinal surgery
.
Gut
.
2005
;
54
(
2
):
237
41
. .
35.
Schoepfer
AM
,
Safroneeva
E
,
Vavricka
SR
,
Peyrin-Biroulet
L
,
Mottet
C
.
Treatment of fibrostenotic and fistulizing Crohn’s disease
.
Digestion
.
2012
;
86 Suppl 1
:
23
7
. .
36.
Rieder
F
,
Latella
G
,
Magro
F
,
Yuksel
ES
,
Higgins
PD
,
Di Sabatino
A
,
European Crohn’s and colitis organisation topical review on prediction, diagnosis and management of fibrostenosing Crohn’s disease
.
J Crohns Colitis
.
2016
;
10
(
8
):
873
85
.
37.
Bettenworth
D
,
Lopez
R
,
Hindryckx
P
,
Levesque
BG
,
Rieder
F
.
Heterogeneity in endoscopic treatment of Crohn’s disease-associated strictures: an international inflammatory bowel disease specialist survey
.
J Gastroenterol
.
2016
;
51
(
10
):
939
48
. .
38.
Zhang
YZ
,
Li
YY
.
Inflammatory bowel disease: pathogenesis
.
World J Gastroenterol
.
2014
;
20
(
1
):
91
9
. .
39.
Guan
Q
.
A comprehensive review and update on the pathogenesis of inflammatory bowel disease
.
J Immunol Res
.
2019
;
2019
:
7247238
. .
40.
Choy
MC
,
Visvanathan
K
,
De Cruz
P
.
An overview of the innate and adaptive immune system in inflammatory bowel disease
.
Inflamm Bowel Dis
.
2017
;
23
(
1
):
2
13
. .
41.
Ueno
A
,
Jeffery
L
,
Kobayashi
T
,
Hibi
T
,
Ghosh
S
,
Jijon
H
.
Th17 plasticity and its relevance to inflammatory bowel disease
.
J Autoimmun
.
2018
;
87
:
38
49
. .
42.
Rieder
F
,
Fiocchi
C
,
Rogler
G
.
Mechanisms, management, and treatment of fibrosis in patients with inflammatory bowel diseases
.
Gastroenterology
.
2017
;
152
(
2
):
340
50.e6
. .
43.
Rieder
F
,
Fiocchi
C
.
Intestinal fibrosis in inflammatory bowel disease: current knowledge and future perspectives
.
J Crohns Colitis
.
2008
;
2
(
4
):
279
90
. .
44.
Chang
CW
,
Wong
JM
,
Tung
CC
,
Shih
IL
,
Wang
HY
,
Wei
SC
.
Intestinal stricture in Crohn’s disease
.
Intest Res
.
2015
;
13
(
1
):
19
26
. .
45.
Pinchuk
IV
,
Mifflin
RC
,
Saada
JI
,
Powell
DW
.
Intestinal mesenchymal cells
.
Curr Gastroenterol Rep
.
2010
;
12
(
5
):
310
8
. .
46.
Vaday
GG
,
Hershkoviz
R
,
Rahat
MA
,
Lahat
N
,
Cahalon
L
,
Lider
O
.
Fibronectin-bound TNF-alpha stimulates monocyte matrix metalloproteinase-9 expression and regulates chemotaxis
.
J Leukoc Biol
.
2000
;
68
(
5
):
737
47
.
47.
Munger
JS
,
Harpel
JG
,
Gleizes
PE
,
Mazzieri
R
,
Nunes
I
,
Rifkin
DB
.
Latent transforming growth factor-beta: structural features and mechanisms of activation
.
Kidney Int
.
1997
;
51
(
5
):
1376
82
. .
48.
Wells
RG
.
The role of matrix stiffness in regulating cell behavior
.
Hepatology
.
2008
;
47
(
4
):
1394
400
. .
49.
Johnson
LA
,
Luke
A
,
Sauder
K
,
Moons
DS
,
Horowitz
JC
,
Higgins
PD
.
Intestinal fibrosis is reduced by early elimination of inflammation in a mouse model of IBD: impact of a “Top-Down” approach to intestinal fibrosis in mice
.
Inflamm Bowel Dis
.
2012
;
18
(
3
):
460
71
. .
50.
Johnson
LA
,
Rodansky
ES
,
Haak
AJ
,
Larsen
SD
,
Neubig
RR
,
Higgins
PD
.
Novel Rho/MRTF/SRF inhibitors block matrix-stiffness and TGF-β-induced fibrogenesis in human colonic myofibroblasts
.
Inflamm Bowel Dis
.
2014
;
20
(
1
):
154
65
. .
51.
Li
C
,
Kuemmerle
JF
.
Mechanisms that mediate the development of fibrosis in patients with Crohn’s disease
.
Inflamm Bowel Dis
.
2014
;
20
(
7
):
1250
8
. .
52.
Martinon
F
,
Tschopp
J
.
NLRs join TLRs as innate sensors of pathogens
.
Trends Immunol
.
2005
;
26
(
8
):
447
54
. .
53.
Stone
JH
,
Zen
Y
,
Deshpande
V
.
IgG4-related disease
.
N Engl J Med
.
2012
;
366
(
6
):
539
51
. .
54.
Wallace
ZS
,
Stone
JH
.
An update on IgG4-related disease
.
Curr Opin Rheumatol
.
2015
;
27
(
1
):
83
90
. .
55.
Mahajan
VS
,
Mattoo
H
,
Deshpande
V
,
Pillai
SS
,
Stone
JH
.
IgG4-related disease
.
Annu Rev Pathol
.
2014
;
9
:
315
47
. .
56.
Aalberse
RC
,
Stapel
SO
,
Schuurman
J
,
Rispens
T
.
Immunoglobulin G4: an odd antibody
.
Clin Exp Allergy
.
2009
;
39
(
4
):
469
77
. .
57.
Shiokawa
M
,
Kodama
Y
,
Sekiguchi
K
,
Kuwada
T
,
Tomono
T
,
Kuriyama
K
,
Laminin 511 is a target antigen in autoimmune pancreatitis
.
Sci Transl Med
.
2018
;
10
(
453
):
eaaq0997
.
58.
Hubers
LM
,
Vos
H
,
Schuurman
AR
,
Erken
R
,
Oude Elferink
RP
,
Burgering
B
,
Annexin A11 is targeted by IgG4 and IgG1 autoantibodies in IgG4-related disease
.
Gut
.
2018
;
67
(
4
):
728
35
.
59.
Perugino
CA
,
AlSalem
SB
,
Mattoo
H
,
Della-Torre
E
,
Mahajan
V
,
Ganesh
G
,
Identification of galectin-3 as an autoantigen in patients with IgG(4)-related disease
.
J Allergy Clin Immunol
.
2019
;
143
(
2
):
736
45.e6
.
60.
Takeuchi
A
,
Saito
T
.
CD4 CTL, a cytotoxic subset of CD4(+) T cells, their differentiation and function
.
Front Immunol
.
2017
;
8
:
194
.
61.
Perugino
CA
,
Stone
JH
.
IgG4-related disease: an update on pathophysiology and implications for clinical care
.
Nat Rev Rheumatol
.
2020
;
16
(
12
):
702
14
. .
62.
Liu
C
,
Zhang
P
,
Zhang
W
.
Immunological mechanism of IgG4-related disease
.
J Transl Autoimmun
.
2020
;
3
:
100047
. .
63.
Mattoo
H
,
Mahajan
VS
,
Maehara
T
,
Deshpande
V
,
Della-Torre
E
,
Wallace
ZS
,
Clonal expansion of CD4(+) cytotoxic T lymphocytes in patients with IgG4-related disease
.
J Allergy Clin Immunol
.
2016
;
138
(
3
):
825
38
.
64.
Della-Torre
E
,
Bozzalla-Cassione
E
,
Sciorati
C
,
Ruggiero
E
,
Lanzillotta
M
,
Bonfiglio
S
,
A CD8α- subset of CD4+SLAMF7+ cytotoxic T cells is expanded in patients with IgG4-related disease and decreases following glucocorticoid treatment
.
Arthritis Rheumatol
.
2018
;
70
(
7
):
1133
43
.
65.
Mattoo
H
,
Stone
JH
,
Pillai
S
.
Clonally expanded cytotoxic CD4(+) T cells and the pathogenesis of IgG4-related disease
.
Autoimmunity
.
2017
;
50
(
1
):
19
24
.
66.
Chen
Y
,
Lin
W
,
Yang
H
,
Wang
M
,
Zhang
P
,
Feng
R
,
Aberrant expansion and function of follicular helper T cell subsets in IgG4-related disease
.
Arthritis Rheumatol
.
2018
;
70
(
11
):
1853
65
.
67.
Xia
C
,
Liu
C
,
Liu
Y
,
Long
Y
,
Xu
L
,
Liu
C
.
Increased circulating Th1 and Tfh1 cell numbers are associated with disease activity in glucocorticoid-treated patients with IgG4-related disease
.
J Immunol Res
.
2020
;
2020
:
3757015
. .
68.
Donnarumma
T
,
Young
GR
,
Merkenschlager
J
,
Eksmond
U
,
Bongard
N
,
Nutt
SL
,
Opposing development of cytotoxic and follicular helper CD4 T cells controlled by the TCF-1-Bcl6 nexus
.
Cell Rep
.
2016
;
17
(
6
):
1571
83
.
69.
Akiyama
M
,
Yasuoka
H
,
Yamaoka
K
,
Suzuki
K
,
Kaneko
Y
,
Kondo
H
,
Enhanced IgG4 production by follicular helper 2 T cells and the involvement of follicular helper 1 T cells in the pathogenesis of IgG4-related disease
.
Arthritis Res Ther
.
2016
;
18
:
167
.
70.
Kubo
S
,
Nakayamada
S
,
Zhao
J
,
Yoshikawa
M
,
Miyazaki
Y
,
Nawata
A
,
Correlation of T follicular helper cells and plasmablasts with the development of organ involvement in patients with IgG4-related disease
.
Rheumatology
.
2018
;
57
(
3
):
514
24
.
71.
Maehara
T
,
Mattoo
H
,
Mahajan
VS
,
Murphy
SJ
,
Yuen
GJ
,
Ishiguro
N
,
The expansion in lymphoid organs of IL-4(+) BATF(+) T follicular helper cells is linked to IgG4 class switching in vivo
.
Life Sci Alliance
.
2018
;
1
(
1
):
e201800050
.
72.
Mattoo
H
,
Mahajan
VS
,
Della-Torre
E
,
Sekigami
Y
,
Carruthers
M
,
Wallace
ZS
,
De novo oligoclonal expansions of circulating plasmablasts in active and relapsing IgG4-related disease
.
J Allergy Clin Immunol
.
2014
;
134
(
3
):
679
87
.
73.
Khosroshahi
A
,
Carruthers
MN
,
Deshpande
V
,
Unizony
S
,
Bloch
DB
,
Stone
JH
.
Rituximab for the treatment of IgG4-related disease: lessons from 10 consecutive patients
.
Medicine
.
2012
;
91
(
1
):
57
66
. .
74.
Rebours
V
,
Le Baleur
Y
,
Cazals-Hatem
D
,
Stefanescu
C
,
Hentic
O
,
Maire
F
,
Immunoglobulin G4 immunostaining of gastric, duodenal, or colonic biopsies is not helpful for the diagnosis of autoimmune pancreatitis
.
Clin Gastroenterol Hepatol
.
2012
;
10
(
1
):
91
4
.
75.
Wang
Z
,
Zhu
M
,
Luo
C
,
zhen
Y
,
Mu
J
,
Zhang
W
,
High level of IgG4 as a biomarker for a new subset of inflammatory bowel disease
.
Sci Rep
.
2018
;
8
(
1
):
10018
.
76.
Chen
X
,
Sun
W
,
Lin
R
,
Huang
Z
,
Chen
W
.
IgG4+ plasma cell infiltration is correlated with the development of inflammatory bowel disease and can be regulated by TLR-4
.
Int J Clin Exp Pathol
.
2018
;
11
(
9
):
4537
44
.
77.
Topal
F
,
Sarıtaş Yüksel
E
,
Ekinci
N
,
Pekdiker
M
,
Cakalağaoğlu
F
,
Alper
E
,
The prevalence of IgG4-positive plasma cell infiltrates in inflammatory bowel disease patients without autoimmune pancreatitis
.
Turk J Gastroenterol
.
2014
;
25
(
5
):
558
62
.
78.
Wei
L
,
Laurence
A
,
Elias
KM
,
O’Shea
JJ
.
IL-21 is produced by Th17 cells and drives IL-17 production in a STAT3-dependent manner
.
J Biol Chem
.
2007
;
282
(
48
):
34605
10
. .
79.
Della-Torre
E
,
Rigamonti
E
,
Perugino
C
,
Baghai-Sain
S
,
Sun
N
,
Kaneko
N
,
B lymphocytes directly contribute to tissue fibrosis in patients with IgG(4)-related disease
.
J Allergy Clin Immunol
.
2020
;
145
(
3
):
968
81.e14
.
80.
Nakase
H
,
Ishigami
K
.
New paradigm of B-cell biology regarding the elucidation of a new mechanism of tissue fibrosis in IgG(4)-related disease
.
J Allergy Clin Immunol
.
2020
;
145
(
3
):
785
7
.
81.
Klinkhammer
BM
,
Floege
J
,
Boor
P
.
PDGF in organ fibrosis
.
Mol Aspects Med
.
2018
;
62
:
44
62
. .
82.
Trojanowska
M
.
Role of PDGF in fibrotic diseases and systemic sclerosis
.
Rheumatology
.
2008
;
47 Suppl 5
:
v2
4
.
83.
Lighaam
LC
,
Aalberse
RC
,
Rispens
T
.
IgG4-related fibrotic diseases from an immunological perspective: regulators out of control?
Int J Rheumatol
.
2012
;
2012
:
789164
. .
84.
François
A
,
Chatelus
E
,
Wachsmann
D
,
Sibilia
J
,
Bahram
S
,
Alsaleh
G
,
B lymphocytes and B-cell activating factor promote collagen and profibrotic markers expression by dermal fibroblasts in systemic sclerosis
.
Arthritis Res Ther
.
2013
;
15
(
5
):
R168
.
85.
Shi
Y
,
Chen
Z
,
Zhao
Z
,
Yu
Y
,
Fan
H
,
Xu
X
,
IL-21 Induces an imbalance of Th17/Treg cells in moderate-to-severe plaque psoriasis patients
.
Front Immunol
.
2019
;
10
:
1865
.
86.
Sutton
CE
,
Lalor
SJ
,
Sweeney
CM
,
Brereton
CF
,
Lavelle
EC
,
Mills
KH
.
Interleukin-1 and IL-23 induce innate IL-17 production from gammadelta T cells, amplifying Th17 responses and autoimmunity
.
Immunity
.
2009
;
31
(
2
):
331
41
. .
87.
Fouser
LA
,
Wright
JF
,
Dunussi-Joannopoulos
K
,
Collins
M
.
Th17 cytokines and their emerging roles in inflammation and autoimmunity
.
Immunol Rev
.
2008
;
226
:
87
102
. .
88.
Yang
L
,
Anderson
DE
,
Baecher-Allan
C
,
Hastings
WD
,
Bettelli
E
,
Oukka
M
,
IL-21 and TGF-beta are required for differentiation of human T(H)17 cells
.
Nature
.
2008
;
454
(
7202
):
350
2
.
89.
Monteleone
G
,
Monteleone
I
,
Fina
D
,
Vavassori
P
,
Del Vecchio Blanco
G
,
Caruso
R
,
Interleukin-21 enhances T-helper cell type I signaling and interferon-gamma production in Crohn’s disease
.
Gastroenterology
.
2005
;
128
(
3
):
687
94
.
90.
Pallone
F
,
Fina
D
,
Caruso
R
,
Monteleone
G
.
Role of IL-21 in inflammatory bowel disease
.
Expert Rev Clin Immunol
.
2010
;
6
(
4
):
537
41
. .
91.
Park
JH
,
Peyrin-Biroulet
L
,
Eisenhut
M
,
Shin
JI
.
IBD immunopathogenesis: a comprehensive review of inflammatory molecules
.
Autoimmun Rev
.
2017
;
16
(
4
):
416
26
. .
92.
Spolski
R
,
Leonard
WJ
.
IL-21 and T follicular helper cells
.
Int Immunol
.
2010
;
22
(
1
):
7
12
. .
93.
Vogelzang
A
,
McGuire
HM
,
Yu
D
,
Sprent
J
,
Mackay
CR
,
King
C
.
A fundamental role for interleukin-21 in the generation of T follicular helper cells
.
Immunity
.
2008
;
29
(
1
):
127
37
. .
94.
Vinuesa
CG
,
Linterman
MA
,
Goodnow
CC
,
Randall
KL
.
T cells and follicular dendritic cells in germinal center B-cell formation and selection
.
Immunol Rev
.
2010
;
237
(
1
):
72
89
. .
95.
Kerfoot
SM
,
Yaari
G
,
Patel
JR
,
Johnson
KL
,
Gonzalez
DG
,
Kleinstein
SH
,
Germinal center B cell and T follicular helper cell development initiates in the interfollicular zone
.
Immunity
.
2011
;
34
(
6
):
947
60
.
96.
Tangye
SG
,
Ma
CS
.
Regulation of the germinal center and humoral immunity by interleukin-21
.
J Exp Med
.
2020
;
217
(
1
):
e20191638
. .
97.
Kuchen
S
,
Robbins
R
,
Sims
GP
,
Sheng
C
,
Phillips
TM
,
Lipsky
PE
,
Essential role of IL-21 in B cell activation, expansion, and plasma cell generation during CD4+ T cell-B cell collaboration
.
J Immunol
.
2007
;
179
(
9
):
5886
96
.
98.
Linterman
MA
,
Beaton
L
,
Yu
D
,
Ramiscal
RR
,
Srivastava
M
,
Hogan
JJ
,
IL-21 acts directly on B cells to regulate Bcl-6 expression and germinal center responses
.
J Exp Med
.
2010
;
207
(
2
):
353
63
.
99.
Zotos
D
,
Coquet
JM
,
Zhang
Y
,
Light
A
,
D’Costa
K
,
Kallies
A
,
IL-21 regulates germinal center B cell differentiation and proliferation through a B cell-intrinsic mechanism
.
J Exp Med
.
2010
;
207
(
2
):
365
78
.
100.
Leonard
WJ
,
Wan
CK
.
IL-21 Signaling in immunity
.
F1000Res
.
2016
;
5
:
F1000 Faculty Rev
224
. .
101.
Ferretti
E
,
Di Carlo
E
,
Ognio
E
,
Guarnotta
C
,
Bertoni
F
,
Corcione
A
,
Interleukin-17A promotes the growth of human germinal center derived non-Hodgkin B cell lymphoma
.
Oncoimmunology
.
2015
;
4
(
10
):
e1030560
.
102.
Ferretti
E
,
Ponzoni
M
,
Doglioni
C
,
Pistoia
V
.
IL-17 superfamily cytokines modulate normal germinal center B cell migration
.
J Leukoc Biol
.
2016
;
100
(
5
):
913
8
. .
103.
Ding
Y
,
Li
J
,
Wu
Q
,
Yang
P
,
Luo
B
,
Xie
S
,
IL-17RA is essential for optimal localization of follicular Th cells in the germinal center light zone to promote autoantibody-producing B cells
.
J Immunol
.
2013
;
191
(
4
):
1614
24
.
104.
Maehara
T
,
Moriyama
M
,
Nakashima
H
,
Miyake
K
,
Hayashida
JN
,
Tanaka
A
,
Interleukin-21 contributes to germinal centre formation and immunoglobulin G4 production in IgG4-related dacryoadenitis and sialoadenitis, so-called Mikulicz’s disease
.
Ann Rheum Dis
.
2012
;
71
(
12
):
2011
9
.
105.
Wood
N
,
Bourque
K
,
Donaldson
DD
,
Collins
M
,
Vercelli
D
,
Goldman
SJ
,
IL-21 effects on human IgE production in response to IL-4 or IL-13
.
Cell Immunol
.
2004
;
231
(
1–2
):
133
45
.
106.
Akiyama
M
,
Suzuki
K
,
Yasuoka
H
,
Kaneko
Y
,
Yamaoka
K
,
Takeuchi
T
.
Follicular helper T cells in the pathogenesis of IgG4-related disease
.
Rheumatology
.
2018
;
57
(
2
):
236
45
. .
107.
Kamekura
R
,
Takano
K
,
Yamamoto
M
,
Kawata
K
,
Shigehara
K
,
Jitsukawa
S
,
Cutting edge: a critical role of lesional T follicular helper cells in the pathogenesis of IgG4-related disease
.
J Immunol
.
2017
;
199
(
8
):
2624
9
.
108.
Timmermans
WM
,
van Laar
JA
,
van der Houwen
TB
,
Kamphuis
LS
,
Bartol
SJ
,
Lam
KH
,
B-cell dysregulation in Crohn’s disease is partially restored with infliximab therapy
.
PLoS One
.
2016
;
11
(
7
):
e0160103
.
109.
Noronha
AM
,
Liang
Y
,
Hetzel
JT
,
Hasturk
H
,
Kantarci
A
,
Stucchi
A
,
Hyperactivated B cells in human inflammatory bowel disease
.
J Leukoc Biol
.
2009
;
86
(
4
):
1007
16
.
110.
Son
HJ
,
Lee
H
,
Kim
JH
,
Yu
IK
,
Han
HY
.
Unrecognised IgG4 association in progressively transformed germinal centers of lymph nodes with subsequent full-blown IgG4-related chronic fibrosing pancreatitis: a case report
.
Malays J Pathol
.
2018
;
40
(
1
):
73
8
.
111.
Kojima
M
,
Nakamura
S
,
Miyawaki
S
,
Ohno
Y
,
Sakata
N
,
Masawa
N
.
Progressive transformation of germinal center presenting with histological features of hyaline-vascular type of Castleman’s disease
.
APMIS
.
2005
;
113
(
4
):
288
95
. .
112.
Siddiqi
IN
,
Brynes
RK
,
Grimm
K
,
O’Malley
DP
,
Wang
E
.
Perifollicular granulomatous inflammation in reactive lymph nodes: a possible morphologic marker for IgG4 plasmacytosis
.
J Hematopathol
.
2011
;
4
(
4
):
207
14
. .
113.
Grimm
KE
,
Barry
TS
,
Chizhevsky
V
,
Hii
A
,
Weiss
LM
,
Siddiqi
IN
,
Histopathological findings in 29 lymph node biopsies with increased IgG4 plasma cells
.
Mod Pathol
.
2012
;
25
(
3
):
480
91
.
114.
Feng
X
,
Xu
X
,
Wang
Y
,
Zheng
Z
,
Lin
G
.
Ectopic germinal centers and IgG4-producing plasmacytes observed in synovia of HLA-B27+ ankylosing spondylitis patients with advanced hip involvement
.
Int J Rheumatol
.
2015
;
2015
:
316421
. .
115.
Kumagai
S
,
Ohtani
H
,
Nagai
T
,
Funa
K
,
Hiwatashi
NO
,
Shimosegawa
,
Platelet-derived growth factor and its receptors are expressed in areas of both active inflammation and active fibrosis in inflammatory bowel disease
.
Tohoku J Exp Med
.
2001
;
195
(
1
):
21
33
.
116.
Kurahashi
M
,
Nakano
Y
,
Peri
LE
,
Townsend
JB
,
Ward
SM
,
Sanders
KM
.
A novel population of subepithelial platelet-derived growth factor receptor α-positive cells in the mouse and human colon
.
Am J Physiol Gastrointest Liver Physiol
.
2013
;
304
(
9
):
G823
34
. .
117.
Rajkumar
VS
,
Shiwen
X
,
Bostrom
M
,
Leoni
P
,
Muddle
J
,
Ivarsson
M
,
Platelet-derived growth factor-beta receptor activation is essential for fibroblast and pericyte recruitment during cutaneous wound healing
.
Am J Pathol
.
2006
;
169
(
6
):
2254
65
.
118.
Fumery
M
,
Yzet
C
,
Chatelain
D
,
Yzet
T
,
Brazier
F
,
LeMouel
JP
,
Colonic strictures in inflammatory bowel disease: epidemiology, complications, and management
.
J Crohns Colitis
.
2021
;
15
(
10
):
1766
73
.
119.
Laurain
PA
,
Guillo
L
,
D’Amico
F
,
Netter
P
,
Danese
S
,
Baumann
C
,
Incidence of and risk factors for colorectal strictures in ulcerative colitis: a multicenter study
.
Clin Gastroenterol Hepatol
.
2021
;
19
(
9
):
1899
905.e1
.
120.
Zen
Y
,
Fujii
T
,
Harada
K
,
Kawano
M
,
Yamada
K
,
Takahira
M
,
Th2 and regulatory immune reactions are increased in immunoglobin G4-related sclerosing pancreatitis and cholangitis
.
Hepatology
.
2007
;
45
(
6
):
1538
46
.
121.
Tanaka
A
,
Moriyama
M
,
Nakashima
H
,
Miyake
K
,
Hayashida
JN
,
Maehara
T
,
Th2 and regulatory immune reactions contribute to IgG4 production and the initiation of Mikulicz disease
.
Arthritis Rheum
.
2012
;
64
(
1
):
254
63
.
122.
Heeringa
JJ
,
Karim
AF
,
van Laar
JAM
,
Verdijk
RM
,
Paridaens
D
,
van Hagen
PM
,
Expansion of blood IgG(4)(+) B, T(H)2, and regulatory T cells in patients with IgG(4)-related disease
.
J Allergy Clin Immunol
.
2018
;
141
(
5
):
1831
43.e10
.
123.
Kusuda
T
,
Uchida
K
,
Miyoshi
H
,
Koyabu
M
,
Satoi
S
,
Takaoka
M
,
Involvement of inducible costimulator- and interleukin 10-positive regulatory T cells in the development of IgG4-related autoimmune pancreatitis
.
Pancreas
.
2011
;
40
(
7
):
1120
30
.
124.
Kawamura
E
,
Hisano
S
,
Nakashima
H
,
Takeshita
M
,
Saito
T
.
Immunohistological analysis for immunological response and mechanism of interstitial fibrosis in IgG4-related kidney disease
.
Mod Rheumatol
.
2015
;
25
(
4
):
571
8
. .
125.
Umehara
H
,
Nakajima
A
,
Nakamura
T
,
Kawanami
T
,
Tanaka
M
,
Dong
L
,
IgG4-related disease and its pathogenesis-cross-talk between innate and acquired immunity
.
Int Immunol
.
2014
;
26
(
11
):
585
95
.
126.
Koike
T
.
IgG4-related disease: why high IgG4 and fibrosis?
Arthritis Res Ther
.
2013
;
15
(
1
):
103
. .
127.
Uchida
K
,
Okazaki
K
.
Roles of regulatory T and B cells in IgG4-related disease
.
Curr Top Microbiol Immunol
.
2017
;
401
:
93
114
.
128.
Moussion
C
,
Ortega
N
,
Girard
JP
.
The IL-1-like cytokine IL-33 is constitutively expressed in the nucleus of endothelial cells and epithelial cells in vivo: a novel “alarmin”?
PLoS One
.
2008
;
3
(
10
):
e3331
.
129.
Nunes
T
,
Bernardazzi
C
,
de Souza
HS
.
Interleukin-33 and inflammatory bowel diseases: lessons from human studies
.
Mediators Inflamm
.
2014
;
2014
:
423957
. .
130.
Pastorelli
L
,
De Salvo
C
,
Vecchi
M
,
Pizarro
TT
.
The Role of IL-33 in gut mucosal inflammation
.
Mediators Inflamm
.
2013
;
2013
:
608187
. .
131.
Williams
MA
,
O’Callaghan
A
,
Corr
SC
.
IL-33 and IL-18 in inflammatory bowel disease etiology and microbial interactions
.
Front Immunol
.
2019
;
10
:
1091
. .
132.
Hazlett
LD
,
McClellan
SA
,
Barrett
RP
,
Huang
X
,
Zhang
Y
,
Wu
M
,
IL-33 shifts macrophage polarization, promoting resistance against pseudomonas aeruginosa keratitis
.
Invest Ophthalmol Vis Sci
.
2010
;
51
(
3
):
1524
32
.
133.
He
R
,
Yin
H
,
Yuan
B
,
Liu
T
,
Luo
L
,
Huang
P
,
IL-33 improves wound healing through enhanced M2 macrophage polarization in diabetic mice
.
Mol Immunol
.
2017
;
90
:
42
9
.
134.
Mosser
DM
,
Edwards
JP
.
Exploring the full spectrum of macrophage activation
.
Nat Rev Immunol
.
2008
;
8
(
12
):
958
69
. .
135.
Steinbach
EC
,
Plevy
SE
.
The role of macrophages and dendritic cells in the initiation of inflammation in IBD
.
Inflamm Bowel Dis
.
2014
;
20
(
1
):
166
75
. .
136.
Edwards
JP
,
Zhang
X
,
Frauwirth
KA
,
Mosser
DM
.
Biochemical and functional characterization of three activated macrophage populations
.
J Leukoc Biol
.
2006
;
80
(
6
):
1298
307
. .
137.
Matta
BM
,
Turnquist
HR
.
Expansion of regulatory T cells in vitro and in vivo by IL-33
.
Methods Mol Biol
.
2016
;
1371
:
29
41
. .
138.
Watanabe
T
,
Minaga
K
,
Kamata
K
,
Kudo
M
,
Strober
W
.
Mechanistic insights into autoimmune pancreatitis and IgG4-related disease
.
Trends Immunol
.
2018
;
39
(
11
):
874
89
. .
139.
Furukawa
S
,
Moriyama
M
,
Miyake
K
,
Nakashima
H
,
Tanaka
A
,
Maehara
T
,
Interleukin-33 produced by M2 macrophages and other immune cells contributes to Th2 immune reaction of IgG4-related disease
.
Sci Rep
.
2017
;
7
(
1
):
42413
.
140.
Rogler
G
.
Pathogenesis of strictures in ulcerative colitis: a field to explore
.
Digestion
.
2011
;
84
(
1
):
10
1
. .
141.
Ebbo
M
,
Grados
A
,
Samson
M
,
Groh
M
,
Loundou
A
,
Rigolet
A
,
Long-term efficacy and safety of rituximab in IgG4-related disease: data from a French nationwide study of thirty-three patients
.
PLoS One
.
2017
;
12
(
9
):
e0183844
.
142.
Carruthers
MN
,
Topazian
MD
,
Khosroshahi
A
,
Witzig
TE
,
Wallace
ZS
,
Hart
PA
,
Rituximab for IgG4-related disease: a prospective, open-label trial
.
Ann Rheum Dis
.
2015
;
74
(
6
):
1171
7
.
143.
Du
FH
,
Mills
EA
,
Mao-Draayer
Y
.
Next-generation anti-CD20 monoclonal antibodies in autoimmune disease treatment
.
Auto Immun Highlights
.
2017
;
8
(
1
):
12
. .
144.
Varma
P
,
Falconer
J
,
Aga
A
,
Prince
HM
,
Pianko
S
.
Rituximab-induced Crohn’s disease
.
Scand J Gastroenterol
.
2017
;
52
(
5
):
606
8
. .
145.
Morita
K
,
Shibano
T
,
Maekawa
K
,
Hattori
M
,
Hida
N
,
Nakamura
S
,
Crohn’s disease following rituximab treatment in a patient with refractory nephrotic syndrome
.
CEN Case Rep
.
2019
;
8
(
1
):
55
60
.
146.
Shankar
U
,
Vijayasekar
K
,
Bansal
R
,
Walfish
A
.
Treatment of rituximab-induced Crohn’s disease with Ustekinumab induction and long-term maintenance of remission
.
Inflamm Bowel Dis
.
2020
;
26
(
1
):
e3
. .
147.
Huang
H
,
Benoist
C
,
Mathis
D
.
Rituximab specifically depletes short-lived autoreactive plasma cells in a mouse model of inflammatory arthritis
.
Proc Natl Acad Sci U S A
.
2010
;
107
(
10
):
4658
. .
148.
Frerichs
KA
,
Bosman
PWC
,
van Velzen
JF
,
Fraaij
PLA
,
Koopmans
MPG
,
Rimmelzwaan
GF
,
Effect of daratumumab on normal plasma cells, polyclonal immunoglobulin levels, and vaccination responses in extensively pre-treated multiple myeloma patients
.
Haematologica
.
2020
;
105
(
6
):
e302
6
.
149.
Khan
ML
,
Colby
TV
,
Viggiano
RW
,
Fonseca
R
.
Treatment with bortezomib of a patient having hyper IgG4 disease
.
Clin Lymphoma Myeloma Leuk
.
2010
;
10
(
3
):
217
9
.
150.
Ogawa
S
,
Ochi
T
,
Shimada
H
,
Inagaki
K
,
Fujita
I
,
Nii
A
,
Anti-PDGF-B monoclonal antibody reduces liver fibrosis development
.
Hepatol Res
.
2010
;
40
(
11
):
1128
41
.
151.
Li
H
,
Zeitelhofer
M
,
Nilsson
I
,
Liu
X
,
Allan
L
,
Gloria
B
,
Development of monoclonal anti-PDGF-CC antibodies as tools for investigating human tissue expression and for blocking PDGF-CC induced PDGFRα signalling in vivo
.
PloS one
.
2018
;
13
(
7
):
e0201089
.
152.
Silfvast-Kaiser
A
,
Paek
SY
,
Menter
A
.
Anti-IL17 therapies for psoriasis
.
Expert Opin Biol Ther
.
2019
;
19
(
1
):
45
54
. .
153.
Wang
J
,
Bhatia
A
,
Krugliak Cleveland
N
,
Gupta
N
,
Dalal
S
,
Rubin
DT
,
Rapid onset of inflammatory bowel disease after receiving secukinumab infusion
.
ACG Case Rep J
.
2018
;
5
:
e56
.
154.
Fauny
M
,
Moulin
D
,
Amico
F
,
Netter
P
,
Petitpain
N
,
Arnone
D
,
Paradoxical gastrointestinal effects of interleukin-17 blockers
.
Ann Rheum Dis
.
2020
;
79
(
9
):
1132
.
155.
Holm
TL
,
Tornehave
D
,
Søndergaard
H
,
Kvist
PH
,
Sondergaard
BC
,
Hansen
L
,
Evaluating IL-21 as a potential therapeutic target in Crohn’s disease
.
Gastroenterol Res Pract
.
2018
;
2018
:
5962624
.
156.
Wang
Y
,
Jiang
X
,
Zhu
J
,
Dan
Y
,
Zhang
X
,
Wang
X
,
IL-21/IL-21R signaling suppresses intestinal inflammation induced by DSS through regulation of Th responses in lamina propria in mice
.
Sci Rep
.
2016
;
6
(
1
):
31881
.
157.
Monteleone
G
,
Monteleone
I
,
Fina
D
,
Vavassori
P
,
Del Vecchio Blanco
G
,
Caruso
R
,
Interleukin-21 enhances T-helper cell type I signaling and interferon-γ production in Crohn’s disease
.
Gastroenterology
.
2005
;
128
(
3
):
687
94
.
Copyright / Drug Dosage / Disclaimer
Copyright: All rights reserved. No part of this publication may be translated into other languages, reproduced or utilized in any form or by any means, electronic or mechanical, including photocopying, recording, microcopying, or by any information storage and retrieval system, without permission in writing from the publisher.
Drug Dosage: The authors and the publisher have exerted every effort to ensure that drug selection and dosage set forth in this text are in accord with current recommendations and practice at the time of publication. However, in view of ongoing research, changes in government regulations, and the constant flow of information relating to drug therapy and drug reactions, the reader is urged to check the package insert for each drug for any changes in indications and dosage and for added warnings and precautions. This is particularly important when the recommended agent is a new and/or infrequently employed drug.
Disclaimer: The statements, opinions and data contained in this publication are solely those of the individual authors and contributors and not of the publishers and the editor(s). The appearance of advertisements or/and product references in the publication is not a warranty, endorsement, or approval of the products or services advertised or of their effectiveness, quality or safety. The publisher and the editor(s) disclaim responsibility for any injury to persons or property resulting from any ideas, methods, instructions or products referred to in the content or advertisements.