Next Article in Journal
Flt3 Signaling in B Lymphocyte Development and Humoral Immunity
Next Article in Special Issue
Insights into the Black Box of Intra-Amniotic Infection and Its Impact on the Premature Lung: From Clinical and Preclinical Perspectives
Previous Article in Journal
Targeting the SREBP-1/Hsa-Mir-497/SCAP/FASN Oncometabolic Axis Inhibits the Cancer Stem-like and Chemoresistant Phenotype of Non-Small Cell Lung Carcinoma Cells
Previous Article in Special Issue
Respiratory and Intestinal Microbiota in Pediatric Lung Diseases—Current Evidence of the Gut–Lung Axis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Pathophysiological Concepts and Management of Pulmonary Manifestation of Pediatric Inflammatory Bowel Disease

1
Catholic Children’s Hospital Wilhelmstift, 22149 Hamburg, Germany
2
Department of Pediatrics, University Medical Center Rostock, 18057 Rostock, Germany
3
Cardio-Pulmonary Institute (CPI), University of Giessen and Marburg Lung Center (UGMLC), German Center of Lung Research (DZL), Justus-Liebig-University, 35398 Giessen, Germany
4
Department of Pediatrics, University Medical Center Greifswald, 17475 Greifswald, Germany
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(13), 7287; https://doi.org/10.3390/ijms23137287
Submission received: 11 June 2022 / Revised: 28 June 2022 / Accepted: 28 June 2022 / Published: 30 June 2022

Abstract

:
Pulmonary manifestation (PM) of inflammatory bowel disease (IBD) in children is a rare condition. The exact pathogenesis is still unclear, but several explanatory concepts were postulated and several case reports in children were published. We performed a systematic Medline search between April 1976 and April 2022. Different pathophysiological concepts were identified, including the shared embryological origin, “miss-homing” of intestinal based neutrophils and T lymphocytes, inflammatory triggering via certain molecules (tripeptide proline-glycine-proline, interleukin 25), genetic factors and alterations in the microbiome. Most pediatric IBD patients with PM are asymptomatic, but can show alterations in pulmonary function tests and breathing tests. In children, the pulmonary parenchyma is more affected than the airways, leading histologically mainly to organizing pneumonia. Medication-associated lung injury has to be considered in pulmonary symptomatic pediatric IBD patients treated with certain agents (i.e., mesalamine, sulfasalazine or infliximab). Furthermore, the risk of pulmonary embolism is generally increased in pediatric IBD patients. The initial treatment of PM is based on corticosteroids, either inhaled for the larger airways or systemic for smaller airways and parenchymal disease. In summary, this review article summarizes the current knowledge about PM in pediatric IBD patients, focusing on pathophysiological and clinical aspects.

1. Introduction

Inflammatory bowel disease (IBD) has shown an increasing prevalence in recent decades worldwide [1]. In 25% of patients, IBD manifestation occurs before the age of 18 [1]. Under the umbrella term of IBD, three different disease entities are subsumed: Crohn’s disease (CD), ulcerative colitis (UC) and inflammatory bowel disease—unspecified [2]. While CD potentially occurs in any part of the digestive tract from mouth to anus and shows an incontiguous, transmural inflammation, UC usually is an ascending, continuous inflammation beginning at the rectal mucosa. The currently accepted pathophysiological idea comprises a dysregulation of the enteric immune system in genetically predisposed individuals, which results in an inadequate response to commensal enteric bacteria [3]. However, IBD is not only understood as an intestinal, but also as a systemic disease, which can affect different organs, called extraintestinal manifestations (EIM) of IBD [4,5]. In the adult IBD population, approximately 30% develop EIM and a quarter of them even before the onset of IBD [6]. If one other organ beside the gut is affected, the likelihood of suffering from further EIM is significantly increased [7]. The prevalence of EIM in the pediatric population was reported to be from 17% to 28% after 15 years of diagnosis [8,9,10]. While 6% of pediatric patients develop EIM before the diagnosis of IBD, one fourth show EIM in the first 2 to 3 years after the diagnosis [10]. Almost ninety percent of EIM will occur in the first year of IBD course [9]. The most common EIM are arthritis, skin lesions (i.e., erythema nodosa and pyoderma gangrenosum), aphthous stomatitis, uveitis and hepatobiliary diseases (i.e., primary sclerosing cholangitis and hepatitis) [10,11,12]. The pulmonary system as a location of EIM is a rare condition (<1%), interstitial pneumonitis being seen more in UC patients and granulomatous disease being more associated with CD [13,14,15]. While in adults pulmonary manifestation (PM) is generally more detected in UC patients, most of the reported cases in children and adolescents are associated with CD [16]. Forty percent of adult IBD patients show respiratory alterations, which are frequently subclinical and might not be clinically relevant [12]. PM in pediatric IBD patients has rarely been described, mostly by case reports [17]. In a large cohort study of >1600 children with IBD, none showed PM [10]. However, it has to be noted that this number might be underrated, due to the fact that the majority of pediatric IBD patients with PM are asymptomatic. Furthermore, no regular screening for pulmonary disease has been established to date in the pediatric IBD population [18,19].
Two very informative review articles have already been published for either the clinical factors or pathophysiological concepts of PM in IBD [5,16]. Therefore, we aim to combine in our review article the most relevant aspects of clinical decision making in children with PM and the current knowledge about the immunological and molecular environment, which most likely leads to the development of PM.

2. Pathophysiological Concepts

Since the first report of a patient with IBD and pulmonary symptoms in 1976 by Kraft el al., several pathophysiological concepts for PM have been proposed [20].

2.1. Shared Embryological Origin

Most of the described links between pulmonary and intestinal tissue are based on the shared embryological origin in the foregut portion of the endoderm, leading to the possibility of a cross-mucosal interaction between the two organs that compose the human mucosal immune system [21,22]. Although much of this gut–lung interaction is not finally understood, both organs show a similar architecture, being composed of an extensive mucosal surface (with goblet cells and submucosal glands), containing lymphoid tissue (innate and adaptive immunity) and being covered by mucus, commensal bacteria and other antigens [16,23,24,25]. The gut and lung share the ability of a mucosal defense mechanism [17]. For instance, the dendritic cells of the lung are able to upregulate the expression of α4β7-integrine, which results in T lymphocyte (Tc) migration to the gut and therefore a direct communication pathway between the intestine and the lung [26]. Furthermore, due to the similar structure of these two mucosal organs, Papanikolaou et al. hypothesized that circulating autoantibodies, such as perinuclear anti-neutrophil cytoplasmic antibodies (pANCA) or anti-saccharomyces cerevisiae antibodies (ASCA), might lead to similar effects of inflammation in the lung as in the gut tissue [16].

2.2. Innate and Adaptive Immunity

In IBD, the homeostasis of the intestinal mucosal immune system is unbalanced, which results in a dysregulated response to luminal, commensal bacteria and other antigens [3]. The first step towards this dysregulation consists of a leaky intestinal mucus and interepithelial cell connection, which facilitates antigens to translocate through the intestinal epithelium [27,28]. In a second step, dendritic cells recognize these antigens as pathogens and induce a T-cell-mediated immune response [29]. Furthermore, activated macrophages induce the expression of leucocyte adhesion molecules at the endothelial blood vessels via the secretion of cytokines (interleukin 1 (IL-1) and tumor necrosis factor alpha (TNF-α)) [30]. This neutrophilic-mediated inflammation plays a key role in IBD development [5]. Neutrophilic cells (Nc) are generated from myeloid precursor cells in the bone marrow, induced by granulocyte colony stimulating factor and IL-17A and then released to the blood stream [31]. Passing by inflamed tissue, Nc become tethered via P-selectin glycoprotein ligand 1 to selectins (P-selectin and E-selectin) expressed by endothelium cells (EC) and consequently migrate into the inflamed tissue (termed margination and diapedesis) [32]. It was shown that the amount of margination and diapedesis of Nc in the pulmonary vasculature during systemic inflammation is augmented, which might be one reason for the trapping of neutrophils in the lung during active IBD [33]. Furthermore, in contrast to other organs, the neutrophilic emigration in the lung vasculature mainly takes place in smaller capillaries and not in post-capillary venules with a larger diameter. Therefore, the pooling time of Nc in the lung is prolonged [34,35]. Moreover, it was shown that pulmonary permeability is increased in patients with CD, facilitating the emigration of Nc [36]. Yipp et al. were able to demonstrate via confocal pulmonary intravital microscopy that, in bacteriemia, pulmonary Nc detect bacterial endotoxins (lipopolysaccharide) via Toll-like receptor 4 (TLR4) binding, resulting in the cluster of differentiation (CD)11b-dependent crawling of Nc. This finally leads to a quicker response to bacterial pathogens compared to the TLR4-myeloid-differentiation primary response gene 88 (Myd88) activation of macrophages. This activation hence triggers, via nuclear factor kappa B (NF-κB), the subsequent expression of TNF-α, chemokines and other proinflammatory molecules, apart from activation of EC to produce selectins for Nc migration [37]. In IBD patients, Nc become initially primed in the gut mucosa during episodes of disease flare. A subset of the these gut-primed Nc are able to relocate from the abluminal to the luminal membrane (reverse transendothelial migration (rTEM)) [38]. Furthermore, the inflamed gut tissue leads to an expression of IL-6, TNF-α, interferon-γ and vascular endothelial growth factor (VEGF) [39,40,41]. Elevated systemic levels of these factors result in an upregulation of ligands for extravasation of Nc and an increased vascular permeability in lung tissue [5,42]. Therefore, the primed (rTEM) Nc recirculate to the bloodstream and translocate into the respiratory endothelium [5]. In vivo studies reveale, that junctional adhesion molecule C is suppressed in inflamed, hypoxic tissue, leading to an increased rTEM of Nc. In addition, primed Nc change their shape and though become deformable, which results in a prolonged transit time through the pulmonary vasculature [38,43]. Interestingly, the healthy lung serves as an organ of de-priming Nc to stabilize the neutrophile homeostasis. In the injured lung, though, this function is compromised, leading to more primed Nc passing the pulmonary blood pool and therefore more Nc transmigrating via the arterial blood stream into the pulmonary mucosa. This finally leads to an exaggerated neutrophilic-mediated inflammation [44]. Aydin et al. found significantly increased VEGF and TNF-α concentration in lung tissue during experimental colitis in rats [45]. TNF-α not only leads to neutrophile accumulation and enhanced expression of adhesion molecules, but also to the formation of granuloma, resulting in the granulomatous manifestation of IBD in the lung [46]. Overall, it can be stated that the close relationship of the mucosal immune systems of the gut and lung via primed (rTEM) Nc might contribute to PM.
Tc represent a second cell type that exhibits a potential mechanism to promote pulmonary inflammation and subsequent tissue damage in the lung. In healthy organs, Tc tend to migrate to the same tissue, where they were first exposed to their specific antigen [47]. Unlike the specific attraction of gut memory Tc via the interaction of α4β7–integrin on Tc and mucosal addressin cell adhesion molecule 1 on intestinal EC, the migration of these Tc to the pulmonary system is based on an unspecific mechanism [48]. The non-tissue-specific binding of L-selectin/CD62L on Tc to peripheral lymph node addressin on EC and/or α4β1-integrin on Tc to vascular cell adhesion molecule 1 on EC results in the migration of lymphocytes to the bronchus-associated lymphatic tissue (BALT) [49]. It was shown that pulmonary Tc express further non-tissue-specific receptors termed C-C chemokine receptor 3 (CCR3) and C-X-C chemokine receptor 5 (CXCR5), which are both increasingly expressed in the Tc of inflamed intestinal tissue [50,51]. Therefore, memory Tc, which experienced their first contact to a specific antigen in the inflamed intestinal mucosa, carry a high number of CCR3 and CXCR5 and, as a result, can translocate to the BALT. This called “miss-homing” explains the presence of gut memory Tc in lung tissue, but it requires an initiation via a gut-specific stimulus to start the inflammatory process. The necessary antigens might derive from a breakdown of the intestinal mucosal barrier during IBD [52].
IL-25 is expressed by respiratory and intestinal epithelial cells and was shown to play an important role in the development of two inflammatory diseases—IBD and bronchial asthma [53,54,55,56]. Experiments in mice revealed that the neutralization of IL-25 results in a decreased inflammation of the colonic mucosa and diminished levels of pro-inflammatory cytokines, such as IL-1β, IL-6 and TNF-α [57]. However, in contrast, IL-25 levels are decreased in the colonic biopsies of CD patients and furthermore pro-inflammatory IL-13 is suppressed by the binding of IL-25 to a specific receptor at CD14+ monocyte-like cells [58]. Therefore, in IBD patients, IL-25 implies a chimeric pathogenic and protective function [59]. While in UC patients inflammation is trigged via T helper cells 2 (Th2), in CD, the expression of Th1-derived cytokines leads to intestinal inflammation [3]. IL-25 is mainly expressed by Th2 [60]. Therefore, it is not surprising that the pro-inflammatory effect of IL-25 is more evident in UC. IL-25 also plays a role in pulmonary inflammation upon allergen provocation [61]. To our knowledge, a specific link between the expression of IL-25 in PM of IBD was studied yet.

2.3. Further Molecular Concepts

The tripeptide proline-glycine-proline (PGP), formed from collagen by two enzymes (matrix metalloproteinase [MMP] and prolyl endopeptidase [PE]), has a chemotactic function for Nc in the intestine by binding to the chemokine receptor CXCR2 on neutrophils [62]. The expression of MMP and PE is elevated in intestinal inflammation. Therefore, more Nc are being attracted, which are able to express MMP and PE themselves [62]. This subsequently results in a neutrophil-mediated intestinal collagen proteolysis and therefore a vicious circle of neutrophilic inflammation [62]. This was shown earlier for inflammatory diseases of the lung, such as chronic obstructive pulmonary disease and asthma, and though it might be one cause of PM of IBD, to date no studies regarding the concentration of PGP in sputum of IBD patients have been published [63].
The already mentioned damage to the epithelial barrier seen in IBD leads to significant bacteremia [64]. This in turn results in neutrophil accumulation in the lung tissue, which is positively associated with levels of IL-1β and C-C-ligand 2 (CCL2), which play a role in pulmonary inflammatory diseases (i.e., bronchiectasis and chronic bronchitis) [65]. Liu et al. discovered, in murine models, that this neutrophil recruitment in the lung is triggered by the expression of platelet-activating factor receptor (PAFR), which then results in the overexpression of the inflammasome via nucleotide-binding oligomerization domain-containing protein (NOD)-like receptor family pyrin-domain-containing 3 (NLRP3) [66]. Therefore, not the actual bacterial colonization, but the activation of lung-hosted inflammasome might play a role in the development of PM [66]. Furthermore, the correlation between an enhanced expression of PAFR and diminished levels of L-selectin on Nc contributes to the trapping of Nc on the endothelial surface and hence the further activation of the inflammation processes [67].
Liu et al. were further able to demonstrate that inflammatory damage to the mucosa and microvascular endothelial changes, such as increased angiogenesis, adhesion molecule expression for lymphocytes (i.e., intercellular adhesion molecule 1), leukocyte extravasation and decreased endothelial barrier function, are similar in the intestinal and pulmonary mucosa during UC [68]. Thromboxane B2 is increasingly expressed in the lung and colonic tissue of murine UC models, which augments leukocyte recruitment and results in mucosal injury via the activation and aggregation of platelets, VEGF-A, which leads to angiogenesis, and finally inducible nitric oxide synthase (iNOS) enhancing leukocyte adhesion [68].

2.4. Microbiome

It seems inevitable that the intestinal microbiome plays an important role in the disease development of IBD [69]. Microbial exposure during early life not only decreases the probability of developing IBD later in childhood or adulthood, but also of other chronic inflammatory diseases occurring in pediatric patients, such as asthma [70,71]. Studying sensitization to food allergens, Stefka et al. discovered that fewer Clostridia spp. result in a dysregulated homeostasis of the host–commensal relationship via reduced IL-22 expression. This finally leads to the alteration of the epithelial junction integrity and the secretion of epithelial antimicrobial peptides [72]. Olszak et al. were able to demonstrate that invariant natural killer T cells accumulate in the lamina propria of the murine gut as well of the lung tissue after contact with commensal microbes early in life. This accumulation is associated with an elevated expression of chemokine ligand CXCL3, which then leads to more lymphatic migration and consequently to IBD or asthma [73].

2.5. Genetic Factors

The concept of IBD as an “organ barrier disease” might underpin a further connection between two organs with a large mucosal surface—the intestine and the lung. Around 40% of CD patients show disease-associated variants of the caspase recruitment domain-containing protein 15 (CARD15) gene, coding for the intracellular NOD2 receptor [74]. These mutations cannot be stated as alone standing factors for their contribution to IBD development, as the genetic susceptibility involvement is thought to be around 20% [75]. The NOD2 receptor, which is not only present in macrophages and dendritic cells, but also in intestinal epithelial cells, is part of a family of pathogen recognition receptors (PRR), recognizing muramyl dipeptide (MDP) as part of the bacterial cell wall [76,77]. The MDP-NOD2-mediated pathway leads via NF-κB activation and translocation to the nucleus to an enhanced expression of α-defensins. In 2004, Wehkamp et al. first discovered that the mutation of NOD2 in patients with CD results in a diminished expression of these α-defensins leading to a breakdown of the mucosal barrier [78]. Therefore, it can be hypothesized that in IBD patients with PM the NOD2 mutation might not only lead to altered inflammation in the gut, but also on the lung surface, especially because NOD2 mutations were also described in the pathogenesis of another inflammatory pulmonary disease—chronic obstructive pulmonary disease (COPD) [79]. Mutations in the autophagy-related 16-like 1 (ATG16L1) gene lead to a diminished function of Paneth cells, a defective antigen presentation and proinflammatory cytokine secretion in IBD patients [80]. ATG16L1 function in PM was not studied to our knowledge, so a concluding assessment, whether the mutation is another link between the intestine and the lung, cannot be made. While specific expressions of humane leucocyte antigen (HLA) are associated with the development of EIM, such as HLA-A2 and HLA-DR1 in CD and HLA-B27, HLA-B8/DR3 and HLA-B58 in UC, no linkage to HLA-types were found for PM [16,81,82].
Figure 1 summarizes the current knowledge about potential cellular and molecular mechanisms involved in the development of PM in patients with IBD.

3. Clinical Aspects

PM in pediatric IBD patients is, due to low incidence and low awareness in clinical settings, a rarely considered aspect. The most important clinical issues will be discussed here with a molecular emphasize.

3.1. Clinical and Radiological Findings

The severity of PM can vary from asymptomatic (detected only by pulmonary function tests (PFT)) to subclinical and severe [83]. Symptoms can be subdivided into those of the upper and lower respiratory tract, interstitial lung disease and pleural affections [84]. In the adult IBD population, the most affected compartment is the bronchial system leading to bronchiectasis or chronic bronchitis, even being diagnosed post-colectomy in patients with UC [25,85,86,87]. Colectomy might constitute a risk factor for the development of PM, explained by a molecular mimicry mechanism, where the pulmonary system is being attacked by earlier gut-sensitized lymphocytes, which lost their target after colectomy [86,88,89,90]. Adult patients present mainly with dyspnea, hoarseness, stridor or chronic coughing [85]. In several studies, the rates of respiratory symptoms in IBD patients were reported from 44 to 50% [91,92,93]. Over 40% revealed abnormal findings on thoracic high resolution computed tomography (HRCT), mainly centrilobular nodules and bronchial wall thickening [94]. Other frequently detected alterations in HRCT include bronchiectasis, reticulonodular opacities, emphysema and ground-glass alterations [12,95].
In the pediatric population, PM generally remains clinically silent and affects primarily the parenchyma (alveolar and interstitial), with organizing pneumonia being the most commonly described pathology [25,96]. On chest X-rays of patients with suspected interstitial involvement, focal or diffuse peripheral predominant opacities and air bronchogram can be detected [12]. HRCT, which is rarely used in children, shows patchy, asymmetric foci of consolidation in a peripheral or peribronchovascular distribution and ground-glass opacities [12]. Looking at radiographic diagnostics in children, Peradzynska et al. reported that HRCT identified only 1 out of 32 pulmonary asymptomatic patient with bilateral bronchiectasis [97]. In most of the published cases of children with IBD and pulmonary symptomatic PM, X-rays showed alterations (Table 1 and Table 2). If the airway system is affected in children with IBD, the inflammation concentrates on smaller airway compartments, leading to purulent bronchitis [18,86,98]. Adult IBD patients with small airway involvement tend to be younger and the proportion of PM, which occurs before intestinal symptoms, is higher than in those patients with large airway involvement [96]. The difference between pathological findings in HRCT and symptoms in children and adults emphasizes that pulmonary involvement in IBD shows a progressive nature, which hardly leads to clinical issues in childhood, but contributes to morbidity in adults.
Table 1 and Table 2 summarize all 34 case reports of PM in pediatric patients with CD (Table 1) and UC (Table 2) to date. While in the adult population PM is more frequently associated with UC, most of the reported pediatric PM cases were children with CD (74%) [87].

3.2. Pulmonary Function and Breathing Tests

PFT play an important role in monitoring children with chronic diseases of the respiratory system, although they are not yet established for screening for pulmonary involvement in children with IBD on a regular basis. Studies with adult patients reported a compromised lung function in up to 94% of patients with active IBD, particularly of the small airways [91,119,120,121,122,123,124]. While spirometry and plethysmography measurements in pediatric IBD patients showed no significant pathological alterations in a number of studies; the lung clearance index (LCI), as a possible marker of early damage and ventilation inhomogeneity, was significantly decreased [97,125,126,127]. This observation can lead to the hypothesis that early damage of the lung in pediatric IBD patients, which is not yet showing any clinical symptoms, might lead over time to more severe damage in adulthood [125].
A recent study showed alterations in certain parameters of PFT (forced expiratory volume in 1 s (FEV1), forced vital capacity and diffusion capacity of lung for carbon monoxide (DLCO)) between the day of IBD diagnosis and three years after [128]. Contradictory to other studies, Furlano et al. showed that pediatric CD patients have significantly decreased mid- and end-expiratory flow values, but the number of patients with flow values under the norm was generally low [98].
If the impaired gas transfer, measured via DLCO, might be a more reliable parameter to objectify the involvement of pulmonary interstitium, in pediatric IBD patients this is likewise not certain, as several studies revealed that pediatric IBD patients showed decreased DLCO levels, whereas other authors could not find any significant correlation [97,98,125,127,129,130].
NOS2 is induced by inflammatory stimuli, leading to a proinflammatory environment [131]. It was shown that the gene coding for NOS2 (NOS2A) is overexpressed in IBD, leading to increased nitric oxide levels in the colonic and rectal gas of UC patients by the factor 100 [132,133]. Therefore, it can be hypothesized that in IBD functional exhaled nitrite oxide (FeNO) levels might be elevated even earlier than PFT can detect airway inflammation. A study of Gut et al. showed that FeNO was elevated in children with IBD (CD and UC) compared to healthy individuals, independently from disease activity [134]. Three other studies with pediatric CD patients do not support this correlation [97,135,136]. In the adult population, the levels of FeNO seem to be associated with the disease activity of CD and UC [137,138].
Alternative methods of non-invasive monitoring of pulmonary inflammation can be obtained via exhaled breath condensate (EBC) samples. A decreased pH in EBC samples of pediatric CD patients, a sign of an inflammatory process, showed significantly lower levels than in healthy controls and in patients with asthma [136]. Furthermore, the correlation to disease activity converged significancy [136]. Other biomarkers, such as 8-isoprostane, a free-radical induced lipid peroxidation product of arachidonic acid, are elevated due to oxidate stress damage (such as in children with asthma) and were surprisingly lower than in the control group, which might be due to different oxidate stress pathways in CD patients [136,139]. The levels of trefoil factor 2 (TTF2) in EBC, a member of a group of growth-factor-like peptides, which are associated with mucosa barrier protection, were, unlike those in asthma patients, significantly decreased [136]. This is consistent with previous studies, which found lower levels of TTF2 in the intestinal mucosa biopsies of pediatric IBD patients than in non-inflamed mucosa samples [140]. Another study group looked at proinflammatory cytokines in EBC (IL-6, IL-8, IL-1β, and TNF-α) and found elevated levels for all parameters in pulmonary asymptomatic pediatric IBD patients with normal PFT [127]. This shows that monitoring of proinflammatory cytokines in EBC might be a further method to detect early inflammatory progress in lung tissue.
There are two studies looking at bronchial hyperresponsiveness (BHR) in pediatric CD patients using methacholine challenge. In a small cohort, 71% of pulmonary asymptomatic pediatric CD patients showed BHR, with no correlation to disease activity, duration of disease course or treatment [141]. Livnat et al. reported 30% of pediatric CD patients with BHR [135]. Whether subclinical BHR is generally important for evaluation of pediatric IBD patients remains unclear.

3.3. Medication-Associated Lung Injury

Pulmonary symptoms in IBD should not be attributed to PM alone, as they can also be caused by medication [142]. Current medical treatment options for pediatric IBD include—beside exclusive enteral nutrition—5-ASA (mesalamine (MES), sulfasalazine (SUF)), immunosuppressives (i.e., prednisolone (PRD) and tacrolimus), immunomodulators (i.e., azathioprine (AZA) or methotrexate (MTX)) and therapies with biological agents (i.e., infliximab (IFX) and adalimumab (ADA); off-label-use in children: vedolizumab (VED), ustekinumab (UST), and tofacitinib) [143,144]. Medication-associated lung injury in pediatric IBD patients is scarce, but plays an important role in the management of pulmonary symptoms, as it is accountable for most non-infectious lung diseases in IBD patients [145]. While the medications used in IBD treatment usually affect more frequently the parenchyma, PM in adult patients are more likely to affect the airways. Therefore, differentiation between these two entities of lung disease might be more reliable than in childhood, where most PM likewise affect the lung parenchyma [142].
The treatment of IBD with MES and SUF are well recognized to cause lung injury with histopathological correlates, such as bilateral interstitial lymphocytic infiltrates, eosinophilic pneumonia, non-necrotizing pulmonary granulomatosis, bronchiolitis obliterans or fibrosing alveolitis [146,147,148]. MES seems to be associated with less pulmonary side effects than SUF [146].
While 25% of IBD patients receiving MTX showed pulmonary symptoms (i.e., cough, wheezing, and dyspnea), only 2–7% of patients will develop pneumonitis within several weeks of initiation [149,150,151].
Lee et al. studied over 1,000 adult IBD patients and reported six cases with anti-TNF-α-induced lung injury [152]. Before initiating therapy with TNF-α antagonists (i.e., IFX or ADA), it is obligatory to exclude active or latent infection with mycobacterium tuberculosis (TB) with Interferon-Gamma Release Assays and chest X-ray [153]. A cohort study of 873 adult IBD patients revealed 25 TB cases, of whom 76% developed TB despite an initially negative screening for latent TB [154]. Apart from TB, other infectious pulmonary side effects of IFX therapy were reported, almost invariably in adulthood (i.e., atypical mycobacterial infection, invasive fungal infections (such as histoplasmosis, aspergillosis or cryptococcosis), infection with pneumocystis jirovecii and nocardia) [155,156,157,158,159,160,161,162]. Review articles published by Tragiannidis et al. and Toussi et al. summarize the current knowledge regarding infections and particularly invasive fungal infections in children receiving TNF-α antagonists [163,164]. Non-infectious lung injury related to IFX include interstitial pneumonitis, alveolitis and diffuse alveolar hemorrhage, regularly resolving after drug discontinuation [165,166,167,168]. The pathogenesis is still unclear, but it was hypothesized that blocking proinflammatory TNF-α leads to a decreased apoptosis rate of inflammatory cells. These cells consequently remain in the pulmonary parenchyma, leading finally to tissue damage [169]. Recently, a case of a child experiencing VED-associated pulmonary disease was published, probably based on an integrin shift leading to proliferation of CD29+ lymphocytes (β1-integrin), which amplifies lymphocyte homing to the lung [170]. Mitchel et al. reported the case of a child with non-infectious bilateral pneumonia after the initiation of immunosuppressive therapy with UST [171]. To our knowledge, although reports of ADA-induced interstitial lung disease in adult IBD have been published, no pediatric cases have been mentioned to date [172]. A Cochrane analysis looked at fecal microbiota transplantation (FMT) in IBD patients and found some significance to achieve clinical remission in adult UC patients [173]. Two small randomized-control trials in CD patients showed a higher clinical remission rate versus placebo, with no reported adverse events [174]. However, pneumonia, as an adverse event using fecal microbiome transplantation, was reported in one UC patient [175].

3.4. Risk of Pulmonary Embolism

The incidence of venous thromboembolism (VTE) in children with IBD has been indicated to be between 0.6 and 1.9% [176,177]. The risk of pulmonary embolism (PE) is raised in children under the age of 16 in the first 5 years after the diagnosis of IBD [178]. A Danish registry study, looking at 5400 patients under 20 years of age, reported a PE incidence rate of 3.4 per 10,000 person-years in children with IBD compared to 0.6 per 10,000 person-years in the non-IBD cohort [179].
Nylund et al. found a relative risk (RR) of pulmonary embolism in CD of 1.8 in comparison to no-IBD children with hospitals stays, whereas UC patients did not show a significant difference [180]. The pathophysiology behind the raised risk of VTE in IBD is not well understood, but a number of alterations of the coagulation cascade and the fibrinolytic system were recognized [181]. Furthermore, persisting inflammation contributes to coagulation via cytokines, such as TNF-α, IL-6, IL-1β or C-reactive protein, which induce the coagulation cascade and inhibit anticoagulation pathways [182,183,184,185]. Recommendations for thromboprophylaxis in children with acute severe colitis and certain risk factors (i.e., smoking, oral contraceptives, complete immobilization and known thrombotic disorder) were implemented [186]. The current pediatric CD guideline of the European Crohn´s and Colitis Organisation (ECCO) and the European Society of Paediatric Gastroenterology, Hepatology and Nutrition (ESPGHAN) do not comment on thromboprophylaxis [143]. In a recently published article by the large European-based pediatric IBD registry named Paediatric Inflammatory Bowel Diseases network for Safety, Efficacy, Treatment and Quality improvement of care (PIBD-SETQuality), it was stated that venous thromboprophylaxis should be considered in all hospitalized children with moderate-to-severe CD with at least one additional VTE risk factor [187].

3.5. Treatment

Treatment options for PM in IBD depend on the localization and severity. While patients with upper airway inflammation, which can lead to severe airway obstruction (i.e., subglottic stenosis), should be treated with intravenous steroids, lower airway inflammation can be treated with inhaled glucocorticoids [87,90]. If upper airway inflammation is refractory to steroid treatment, interventional bronchoscopy can be performed (i.e., laser beam, balloon dilatation or stent placement) [16]. Furthermore, when PM is noticed, the escalation of IBD therapy is recommended [188]. In bronchiectasis, rare in children, antibiotic regimes and bronchial toilet should be applied [16]. The parenchymal involvement of PM, which is mostly seen in children, is best targeted with oral glucocorticoids [96,189]. When glucocorticoid therapy is not sufficient (which might be the case in one third of patients), IFX can be administered, if not yet part of the therapy management [190]. Krishan et al. reported three children with PM, who dramatically improved after the initiation of IFX treatment [105]. In cases of bronchiolitis, obliterans macrolides resulted in clinical improvement [16,191]. When medication-induced lung injury is considered, the offending drug has to be stopped and replaced [192].

4. Conclusions

The PM of IBD in children is an underestimated clinical problem. Most of the reported symptomatic pediatric cases covered CD patients. Current pathophysiological concepts of PM consist in the shared embryological origin of the gut and lung, a close connection between the pulmonary and intestinal immune system, the similar expression of distinctive proinflammatory molecules, genetic susceptibility and aspects of the microbiome. PM in children stays mostly silent, affecting more often the parenchyma than in the adult population. Due to contradictory study results in pediatric IBD patients, PFT are currently not playing a role yet in screening for PM, but measurement of LCI might show a promising way to detect early lung damage. Importantly, interstitial lung disease in children with IBD can also be caused by medications, such as mesalamine or biological therapies. PM can be generally treated with corticosteroids, either inhaled for the larger airways, or systemically for smaller airway involvement and parenchymal inflammation. When corticosteroids show no sufficient effect, IFX can be applied. Screening for PM in children with IBD should be considered a reasonable tool to prevent progressive lung damage in adulthood.

5. Methods

A systemic Medline search using the terms “lung”, “pulmonary”, “inflammatory bowel disease”, “Crohn’s disease”, “ulcerative colitis”, “IBD unclassified”, “pediatrics”, and “child” was performed. The databases searched included Cochrane, Embase and PubMed from April 1976 to April 2022. Articles not published in English language were excluded.

Author Contributions

Project idea, C.-M.C. and J.D.; conceptualization, F.S. and J.D.; supervision and project administration, J.D.; systematic literature search, F.S. and J.D.; methodology, F.S. and J.D.; formal analyses and interpretation of data, F.S. and J.D.; writing—original draft preparation, F.S.; writing—review and editing, C.-M.C. and J.D. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The authors thank Johannes Knierer (Catholic Children’s Hospital Wilhelmstift, Hamburg, Germany) for support in creating the figures. The authors acknowledge support for the Article Processing Charge (APC) from the German Research Foundation (DFG) and the Open Access Publication Fund of the University of Greifswald.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Benchimol, E.I.; Fortinsky, K.J.; Gozdyra, P.; Van Den Heuvel, M.; Van Limbergen, J.; Griffiths, A.M. Epidemiology of Pediatric Inflammatory Bowel Disease: A Systematic Review of International Trends. Inflamm. Bowel Dis. 2011, 17, 423–439. [Google Scholar] [CrossRef] [PubMed]
  2. Bousvaros, A.; Antonioli, D.; Colletti, R.; Dubinsky, M.; Glickman, J.; Gold, B.; Griffiths, A.; Jevon, G.; Higuchi, L.; Hyams, J.; et al. Differentiating Ulcerative Colitis from Crohn Disease in Children and Young Adults: Report of a Working Group of the North American Society for Pediatric Gastroenterology, Hepatology, and Nutrition and the Crohn’s and Colitis Foundation of America. J. Pediatr. Gastroenterol. Nutr. 2007, 44, 653–674. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Baumgart, D.C.; Carding, S.R. Inflammatory Bowel Disease: Cause and Immunobiology. Lancet 2007, 369, 1627–1640. [Google Scholar] [CrossRef]
  4. Hedin, C.R.H.; Vavricka, S.R.; Stagg, A.J.; Schoepfer, A.; Raine, T.; Puig, L.; Pleyer, U.; Navarini, A.; Van Der Meulen-De Jong, A.E.; Maul, J.; et al. The Pathogenesis of Extraintestinal Manifestations: Implications for IBD Research, Diagnosis, and Therapy. J. Crohn’s Colitis 2019, 13, 541–554. [Google Scholar] [CrossRef]
  5. Mateer, S.W.; Maltby, S.; Marks, E.; Foster, P.S.; Horvat, J.C.; Hansbro, P.M.; Keely, S. Potential Mechanisms Regulating Pulmonary Pathology in Inflammatory Bowel Disease. J. Leukoc. Biol. 2015, 98, 727–737. [Google Scholar] [CrossRef] [Green Version]
  6. Vavricka, S.R.; Rogler, G.; Gantenbein, C.; Spoerri, M.; Vavricka, M.P.; Navarini, A.A.; French, L.E.; Safroneeva, E.; Fournier, N.; Straumann, A.; et al. Chronological Order of Appearance of Extraintestinal Manifestations Relative to the Time of IBD Diagnosis in the Swiss Inflammatory Bowel Disease Cohort. Inflamm. Bowel Dis. 2015, 21, 1794–1800. [Google Scholar] [CrossRef]
  7. Veloso, F.T.; Carvalho, J.; Magro, F. Immune-Related Systemic Manifestations of Inflammatory Bowel Disease: A Prospective Study of 792 Patients. J. Clin. Gastroenterol. 1996, 23, 29–34. [Google Scholar] [CrossRef]
  8. Greuter, T.; Bertoldo, F.; Rechner, R.; Straumann, A.; Biedermann, L.; Zeitz, J.; Misselwitz, B.; Scharl, M.; Rogler, G.; Safroneeva, E.; et al. Extraintestinal Manifestations of Pediatric Inflammatory Bowel Disease: Prevalence, Presentation, and Anti-TNF Treatment. J. Pediatr. Gastroenterol. Nutr. 2017, 65, 200–206. [Google Scholar] [CrossRef] [Green Version]
  9. Dotson, J.L.; Hyams, J.S.; Markowitz, J.; Leleiko, N.S.; MacK, D.R.; Evans, J.S.; Pfefferkorn, M.D.; Griffiths, A.M.; Otley, A.R.; Bousvaros, A.; et al. Extraintestinal Manifestations of Pediatric Inflammatory Bowel Disease and Their Relation to Disease Type and Severity. J. Pediatr. Gastroenterol. Nutr. 2010, 51, 140–145. [Google Scholar] [CrossRef]
  10. Jose, F.A.; Garnett, E.A.; Vittinghoff, E.; Ferry, G.D.; Winter, H.S.; Baldassana, R.N.; Kirschner, B.S.; Cohen, S.A.; Gold, B.D.; Abramson, O.; et al. Development of Extraintestinal Manifestations in Pediatric Patients with Inflammatory Bowel Disease. Inflamm. Bowel Dis. 2009, 15, 63–68. [Google Scholar] [CrossRef] [Green Version]
  11. Al-Binali, A.M.; Scott, B.; Al-Garni, A.; Montgomery, M.; Robertson, M. Granulomatous Pulmonary Disease in a Child: An Unusual Presentation of Crohn’s Disease. Pediatr. Pulmonol. 2003, 36, 76–80. [Google Scholar] [CrossRef] [PubMed]
  12. Cozzi, D.; Moroni, C.; Addeo, G.; Danti, G.; Lanzetta, M.M.; Cavigli, E.; Falchini, M.; Marra, F.; Piccolo, C.L.; Brunese, L.; et al. Radiological Patterns of Lung Involvement in Inflammatory Bowel Disease. Gastroenterol. Res. Pract. 2018, 2018, 5697846. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Eliadou, E.; Moleiro, J.; Ribaldone, D.G.; Astegiano, M.; Rothfuss, K.; Taxonera, C.; Ghalim, F.; Carbonnel, F.; Verstockt, B.; Festa, S.; et al. Interstitial and Granulomatous Lung Disease in Inflammatory Bowel Disease Patients. J. Crohn’s Colitis 2020, 14, 480–489. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Inoue, K.; Kusunoki, T.; Fujii, T. Organizing Pneumonia as an Extraintestinal Manifestation of Crohn’s Disease in a Child. Pediatr. Pulmonol. 2017, 52, e64–e66. [Google Scholar] [CrossRef]
  15. Rogers, B.H.G.; Clark, L.M.; Kirsner, J.B. The Epidemiologic and Demographic Characteristics of Inflammatory Bowel Disease: An Analysis of a Computerized File of 1400 Patients. J. Chronic Dis. 1971, 24, 743–773. [Google Scholar] [CrossRef]
  16. Papanikolaou, I.; Kagouridis, M.; Papiris, S. Patterns of Airway Involvement in Inflammatory Bowel Diseases. World J. Gastrointest. Pathophysiol. 2014, 5, 560. [Google Scholar] [CrossRef] [PubMed]
  17. Vadlamudi, N.B.; Navaneethan, U.; Thame, K.A.; Kelly, D.R.; Dimmitt, R.A.; Harris, W.T. Crohn’s Disease with Pulmonary Manifestations in Children: 2 Case Reports and Review of the Literature. J. Crohns Colitis 2013, 7, e85–e92. [Google Scholar] [CrossRef]
  18. Taylor, M.A.; Zhou, H.; Dansie, D.M.; Short, S. Pulmonary Manifestations of Inflammatory Bowel Disease in Children. Cureus 2020, 12, e11369. [Google Scholar] [CrossRef]
  19. Barfield, E.; Deshmukh, F.; Slighton, E.; Lentine, J.; Lu, Y.; Ma, X.; Christos, P.J.; Sockolow, R.E.; Loughlin, G.; Pillai, S. Pulmonary Manifestations in Adolescents With Inflammatory Bowel Disease. Clin. Pediatr. 2020, 59, 573–579. [Google Scholar] [CrossRef]
  20. Kraft, S.C.; Earle, R.H.; Roesler, M.; Esterly, J.R. Unexplained Bronchopulmonary Disease With Inflammatory Bowel Disease. Arch. Intern. Med. 1976, 136, 454–459. [Google Scholar] [CrossRef]
  21. Zhang, M.; Zhou, Y.; Li, H.; Peng, Y.; Qiu, P.; Shi, X.; Pan, X.; Liu, J. COVID-19: Gastrointestinal Symptoms from the View of Gut-Lung Axis. Eur. J. Gastroenterol. Hepatol. 2021, 33, 610–612. [Google Scholar] [CrossRef] [PubMed]
  22. Shu, W.; Lu, M.M.; Zhang, Y.; Tucker, P.W.; Zhou, D.; Morrisey, E.E. Foxp2 and Foxp1 Cooperatively Regulate Lung and Esophagus Development. Development 2007, 134, 1991–2000. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Keely, S.; Talley, N.J.; Hansbro, P.M. Pulmonary-Intestinal Cross-Talk in Mucosal Inflammatory Disease. Mucosal Immunol. 2012, 5, 7–18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Mestecky, J. The Common Mucosal Immune System and Current Strategies for Induction of Immune Responses in External Secretions. J. Clin. Immunol. 1987, 7, 265–276. [Google Scholar] [CrossRef]
  25. Gut, G.; Sivan, Y. Respiratory Involvement in Children with Inflammatory Bowel Disease. Pediatr. Allergy Immunol. Pulmonol. 2011, 24, 197–206. [Google Scholar] [CrossRef]
  26. Ruane, D.; Brane, L.; Reis, B.S.; Cheong, C.; Poles, J.; Do, Y.; Zhu, H.; Velinzon, K.; Choi, J.H.; Studt, N.; et al. Lung Dendritic Cells Induce Migration of Protective T Cells to the Gastrointestinal Tract. J. Exp. Med. 2013, 210, 1871–1888. [Google Scholar] [CrossRef] [Green Version]
  27. Söderholm, J.D.; Olaison, G.; Peterson, K.H.; Franzén, L.E.; Lindmark, T.; Wirén, M.; Tagesson, C.; Sjódahl, R. Augmented Increase in Tight Junction Permeability by Luminal Stimuli in the Non-Inflamed Ileum of Crohn’s Disease. Gut 2002, 50, 307–313. [Google Scholar] [CrossRef] [Green Version]
  28. Johansson, M.E.V.; Gustafsson, J.K.; Holmen-Larsson, J.; Jabbar, K.S.; Xia, L.; Xu, H.; Ghishan, F.K.; Carvalho, F.A.; Gewirtz, A.T.; Sjovall, H.; et al. Bacteria Penetrate the Normally Impenetrable Inner Colon Mucus Layer in Both Murine Colitis Models and Patients with Ulcerative Colitis. Gut 2014, 63, 281–291. [Google Scholar] [CrossRef]
  29. Bernardo, D.; Chaparro, M.; Gisbert, J.P. Human Intestinal Dendritic Cells in Inflammatory Bowel Diseases. Mol. Nutr. Food Res. 2018, 62, e1700931. [Google Scholar] [CrossRef]
  30. Charo, I.F.; Ransohoff, R.M. The Many Roles of Chemokines and Chemokine Receptors in Inflammation. N. Engl. J. Med. 2006, 354, 610–621. [Google Scholar] [CrossRef]
  31. Kolaczkowska, E.; Kubes, P. Neutrophil Recruitment and Function in Health and Inflammation. Nat. Rev. Immunol. 2013, 13, 159–175. [Google Scholar] [CrossRef] [PubMed]
  32. Ley, K.; Laudanna, C.; Cybulsky, M.I.; Nourshargh, S. Getting to the Site of Inflammation: The Leukocyte Adhesion Cascade Updated. Nat. Rev. Immunol. 2007, 7, 678–689. [Google Scholar] [CrossRef] [PubMed]
  33. Haslett, C.; Worthen, G.S.; Giclas, P.C.; Morrison, D.C.; Henson, J.E.; Henson, P.M. The Pulmonary Vascular Sequestration of Neutrophils in Endotoxemia Is Initiated by an Effect of Endotoxin on the Neutrophil in the Rabbit. Am. Rev. Respir. Dis. 1987, 136, 9–18. [Google Scholar] [CrossRef] [PubMed]
  34. Lien, D.C.; Wagner, W.W.; Capen, R.L.; Haslett, C.; Hanson, W.L.; Hofmeister, S.E.; Henson, P.M.; Worthen, G.S. Physiological Neutrophil Sequestration in the Lung: Visual Evidence for Localization in Capillaries. J. Appl. Physiol. 1987, 62, 1236–1243. [Google Scholar] [CrossRef]
  35. Hogg, J.C.; Coxson, H.O.; Brumwell, M.L.; Beyers, N.; Doerschuk, C.M.; MacNee, W.; Wiggs, B.R. Erythrocyte and Polymorphonuclear Cell Transit Time and Concentration in Human Pulmonary Capillaries. J. Appl. Physiol. 1994, 77, 1795–1800. [Google Scholar] [CrossRef]
  36. Adenis, A.; Colombel, J.F.; Lecouffe, P.; Wallaert, B.; Hecquet, B.; Marchandise, X.; Cortot, A. Increased Pulmonary and Intestinal Permeability in Crohn’s Disease. Gut 1992, 33, 678–682. [Google Scholar] [CrossRef] [Green Version]
  37. Yipp, B.G.; Kim, J.H.; Lima, R.; Zbytnuik, L.D.; Petri, B.; Swanlund, N.; Ho, M.; Szeto, V.G.; Tak, T.; Koenderman, L.; et al. The Lung Is a Host Defense Niche for Immediate Neutrophil-Mediated Vascular Protection. Sci. Immunol. 2017, 2, eaam8929. [Google Scholar] [CrossRef] [Green Version]
  38. Woodfin, A.; Voisin, M.B.; Beyrau, M.; Colom, B.; Caille, D.; Diapouli, F.M.; Nash, G.B.; Chavakis, T.; Albelda, S.M.; Rainger, G.E.; et al. The Junctional Adhesion Molecule JAM-C Regulates Polarized Transendothelial Migration of Neutrophils in Vivo. Nat. Immunol. 2011, 12, 761–769. [Google Scholar] [CrossRef] [Green Version]
  39. Funderburg, N.T.; Stubblefield Park, S.R.; Sung, H.C.; Hardy, G.; Clagett, B.; Ignatz-Hoover, J.; Harding, C.V.; Fu, P.; Katz, J.A.; Lederman, M.M.; et al. Circulating CD4(+) and CD8(+) T Cells Are Activated in Inflammatory Bowel Disease and Are Associated with Plasma Markers of Inflammation. Immunology 2013, 140, 87–97. [Google Scholar] [CrossRef]
  40. Scaldaferri, F.; Vetrano, S.; Sans, M.; Arena, V.; Straface, G.; Stigliano, E.; Repici, A.; Sturm, A.; Malesci, A.; Panes, J.; et al. VEGF-A Links Angiogenesis and Inflammation in Inflammatory Bowel Disease Pathogenesis. Gastroenterology 2009, 136, 585–595.e5. [Google Scholar] [CrossRef] [Green Version]
  41. Raddatz, D.; Bockemühl, M.; Ramadori, G. Quantitative Measurement of Cytokine MRNA in Inflammatory Bowel Disease: Relation to Clinical and Endoscopic Activity and Outcome. Eur. J. Gastroenterol. Hepatol. 2005, 17, 547–557. [Google Scholar] [CrossRef] [PubMed]
  42. Le Cras, T.D.; Spitzmiller, R.E.; Albertine, K.H.; Greenberg, J.M.; Whitsett, J.A.; Akeson, A.L. VEGF Causes Pulmonary Hemorrhage, Hemosiderosis, and Air Space Enlargement in Neonatal Mice. Am. J. Physiol. Lung Cell. Mol. Physiol. 2004, 287, L134–L142. [Google Scholar] [CrossRef] [PubMed]
  43. Downey, G.P.; Doherty, D.E.; Schwab, B.; Elson, E.L.; Henson, P.M.; Worthen, G.S. Retention of Leukocytes in Capillaries: Role of Cell Size and Deformability. J. Appl. Physiol. 1990, 69, 1767–1778. [Google Scholar] [CrossRef] [PubMed]
  44. Nahum, A.; Chamberlin, W.; Sznajder, J.I.; Hegarty, M. Differential Activation of Mixed Venous and Arterial Neutrophils in Patients with Sepsis Syndrome and Acute Lung Injury. Am. Rev. Respir. Dis. 1991, 143, 1083–1087. [Google Scholar] [CrossRef] [PubMed]
  45. Aydin, B.; Songur, Y.; Songur, N.; Aksu, O.; Senol, A.; Metin Ciris, I.; Sutcu, R. Investigation of Pulmonary Involvement in Inflammatory Bowel Disease in an Experimental Model of Colitis. Korean J. Intern. Med. 2016, 31, 853–859. [Google Scholar] [CrossRef] [Green Version]
  46. Vilcek, J.; Lee, T.H. Tumor Necrosis Factor. New Insights into the Molecular Mechanisms of Its Multiple Actions. J. Biol. Chem. 1991, 266, 7313–7316. [Google Scholar] [CrossRef]
  47. Campbell, D.J.; Butcher, E.C. Brief Definitive Report Rapid Acquisition of Tissue-Specific Homing Phenotypes by CD4 T Cells Activated in Cutaneous or Mucosal Lymphoid Tissues. J. Exp. Med. 2002, 195, 135–141. [Google Scholar] [CrossRef] [Green Version]
  48. von Andrian, U.H.; Mackay, C.R. T-Cell Function and Migration. Two Sides of the Same Coin. N. Engl. J. Med. 2000, 343, 1020–1034. [Google Scholar] [CrossRef]
  49. Xu, B.; Wagner, N.; Pham, L.N.; Magno, V.; Shan, Z.; Butcher, E.C.; Michie, S.A. Lymphocyte Homing to Bronchus-Associated Lymphoid Tissue (BALT) Is Mediated by L-Selectin/PNAd, Alpha4beta1 Integrin/VCAM-1, and LFA-1 Adhesion Pathways. J. Exp. Med. 2003, 197, 1255–1267. [Google Scholar] [CrossRef] [Green Version]
  50. Yuan, Y.H.; Hove, T.; Ten; Olle The, F.; Slors, J.F.M.; Van Deventer, S.J.H.; Te Velde, A.A. Chemokine Receptor CXCR3 Expression in Inflammatory Bowel Disease. Inflamm. Bowel Dis. 2001, 7, 281–286. [Google Scholar] [CrossRef] [Green Version]
  51. Agace, W.W.; Roberts, A.I.; Wu, L.; Greineder, C.; Ebert, E.C.; Parker, C.M. Human Intestinal Lamina Propria and Intraepithelial Lymphocytes Express Receptors Specific for Chemokines Induced by Inflammation. Eur. J. Immunol. 2000, 30, 819–826. [Google Scholar] [CrossRef]
  52. Rojo, Ó.P.; San Román, A.L.; Arbizu, E.A.; Martínez, A.D.L.H.; Sevillano, E.R.; Martínez, A.A. Serum Lipopolysaccharide-Binding Protein in Endotoxemic Patients with Inflammatory Bowel Disease. Inflamm. Bowel Dis. 2007, 13, 269–277. [Google Scholar] [CrossRef] [PubMed]
  53. Haber, A.L.; Biton, M.; Rogel, N.; Herbst, R.H.; Shekhar, K.; Smillie, C.; Burgin, G.; Delorey, T.M.; Howitt, M.R.; Katz, Y.; et al. A Single-Cell Survey of the Small Intestinal Epithelium. Nature 2017, 551, 333–339. [Google Scholar] [CrossRef] [PubMed]
  54. Lloyd, C.M.; Saglani, S. Epithelial Cytokines and Pulmonary Allergic Inflammation. Curr. Opin. Immunol. 2015, 34, 52–58. [Google Scholar] [CrossRef] [PubMed]
  55. Kouzaki, H.; Tojima, I.; Kita, H.; Shimizu, T. Transcription of Interleukin-25 and Extracellular Release of the Protein Is Regulated by Allergen Proteases in Airway Epithelial Cells. Am. J. Respir. Cell Mol. Biol. 2013, 49, 741–750. [Google Scholar] [CrossRef] [Green Version]
  56. Angkasekwinai, P.; Park, H.; Wang, Y.H.; Wang, Y.H.; Seon, H.C.; Corry, D.B.; Liu, Y.J.; Zhu, Z.; Dong, C. Interleukin 25 Promotes the Initiation of Proallergic Type 2 Responses. J. Exp. Med. 2007, 204, 1509–1517. [Google Scholar] [CrossRef] [Green Version]
  57. Reynolds, J.M.; Lee, Y.H.; Shi, Y.; Wang, X.; Angkasekwinai, P.; Nallaparaju, K.C.; Flaherty, S.; Chang, S.H.; Watarai, H.; Dong, C. Interleukin-17B Antagonizes Interleukin-25-Mediated Mucosal Inflammation. Immunity 2015, 42, 692–703. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Caruso, R.; Sarra, M.; Stolfi, C.; Rizzo, A.; Fina, D.; Fantini, M.C.; Pallone, F.; MacDonald, T.T.; Monteleone, G. Interleukin-25 Inhibits Interleukin-12 Production and Th1 Cell-Driven Inflammation in the Gut. Gastroenterology 2009, 136, 2270–2279. [Google Scholar] [CrossRef]
  59. Borowczyk, J.; Shutova, M.; Brembilla, N.C.; Boehncke, W.H. IL-25 (IL-17E) in Epithelial Immunology and Pathophysiology. J. Allergy Clin. Immunol. 2021, 148, 40–52. [Google Scholar] [CrossRef]
  60. Fort, M.M.; Cheung, J.; Yen, D.; Li, J.; Zurawski, S.M.; Lo, S.; Menon, S.; Clifford, T.; Hunte, B.; Lesley, R.; et al. IL-25 Induces IL-4, IL-5, and IL-13 and Th2-Associated Pathologies in Vivo. Immunity 2001, 15, 985–995. [Google Scholar] [CrossRef] [Green Version]
  61. Tamachi, T.; Maezawa, Y.; Ikeda, K.; Kagami, S.I.; Hatano, M.; Seto, Y.; Suto, A.; Suzuki, K.; Watanabe, N.; Saito, Y.; et al. IL-25 Enhances Allergic Airway Inflammation by Amplifying a TH2 Cell-Dependent Pathway in Mice. J. Allergy Clin. Immunol. 2006, 118, 606–614. [Google Scholar] [CrossRef] [PubMed]
  62. Koelink, P.J.; Overbeek, S.A.; Braber, S.; Morgan, M.E.; Henricks, P.A.J.; Roda, M.A.; Verspaget, H.W.; Wolfkamp, S.C.; Te Velde, A.A.; Jones, C.W.; et al. Collagen Degradation and Neutrophilic Infiltration: A Vicious Circle in Inflammatory Bowel Disease. Gut 2014, 63, 578–587. [Google Scholar] [CrossRef] [PubMed]
  63. O’Reilly, P.; Jackson, P.L.; Noerager, B.; Parker, S.; Dransfield, M.; Gaggar, A.; Blalock, J.E. N-Alpha-PGP and PGP, Potential Biomarkers and Therapeutic Targets for COPD. Respir. Res. 2009, 10, 38. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Keely, S.; Campbell, E.L.; Baird, A.W.; Hansbro, P.M.; Shalwitz, R.A.; Kotsakis, A.; McNamee, E.N.; Eltzschig, H.K.; Kominsky, D.J.; Colgan, S.P. Contribution of Epithelial Innate Immunity to Systemic Protection Afforded by Prolyl Hydroxylase Inhibition in Murine Colitis. Mucosal Immunol. 2013, 7, 114–123. [Google Scholar] [CrossRef] [Green Version]
  65. Mateer, S.W.; Mathe, A.; Bruce, J.; Liu, G.; Maltby, S.; Fricker, M.; Goggins, B.J.; Tay, H.L.; Marks, E.; Burns, G.; et al. IL-6 Drives Neutrophil-Mediated Pulmonary Inflammation Associated with Bacteremia in Murine Models of Colitis. Am. J. Pathol. 2018, 188, 1625–1639. [Google Scholar] [CrossRef] [Green Version]
  66. Liu, G.; Mateer, S.W.; Hsu, A.; Goggins, B.J.; Tay, H.; Mathe, A.; Fan, K.; Neal, R.; Bruce, J.; Burns, G.; et al. Platelet Activating Factor Receptor Regulates Colitis-Induced Pulmonary Inflammation through the NLRP3 Inflammasome. Mucosal Immunol. 2019, 12, 862–873. [Google Scholar] [CrossRef]
  67. Mitchell, M.J.; Lin, K.S.; King, M.R. Fluid Shear Stress Increases Neutrophil Activation via Platelet-Activating Factor. Biophys. J. 2014, 106, 2243–2253. [Google Scholar] [CrossRef] [Green Version]
  68. Liu, Y.; Wang, X.Y.; Yang, X.; Jing, S.; Zhu, L.; Gao, S.H. Lung and Intestine: A Specific Link in an Ulcerative Colitis Rat Model. Gastroenterol. Res. Pract. 2013, 2013, 124530. [Google Scholar] [CrossRef] [Green Version]
  69. Chang, J.T. Pathophysiology of Inflammatory Bowel Diseases. N. Engl. J. Med. 2020, 383, 2652–2664. [Google Scholar] [CrossRef]
  70. López-Serrano, P.; Pérez-Calle, J.L.; Pérez-Fernández, M.T.; Fernández-Font, J.M.; Boixeda De Miguel, D.; Fernández-Rodríguez, C.M. Environmental Risk Factors in Inflammatory Bowel Diseases. Investigating the Hygiene Hypothesis: A Spanish Case–Control Study. Scand. J. Gastroenterol. 2010, 45, 1464–1471. [Google Scholar] [CrossRef]
  71. Birzele, L.T.; Depner, M.; Ege, M.J.; Engel, M.; Kublik, S.; Bernau, C.; Loss, G.J.; Genuneit, J.; Horak, E.; Schloter, M.; et al. Environmental and Mucosal Microbiota and Their Role in Childhood Asthma. Allergy 2016, 72, 109–119. [Google Scholar] [CrossRef] [PubMed]
  72. Stefka, A.T.; Feehley, T.; Tripathi, P.; Qiu, J.; McCoy, K.; Mazmanian, S.K.; Tjota, M.Y.; Seo, G.Y.; Cao, S.; Theriault, B.R.; et al. Commensal Bacteria Protect against Food Allergen Sensitization. Proc. Natl. Acad. Sci. USA 2014, 111, 13145–13150. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Olszak, T.; An, D.; Zeissig, S.; Vera, M.P.; Richter, J.; Franke, A.; Glickman, J.N.; Siebert, R.; Baron, R.M.; Kasper, D.L.; et al. Microbial Exposure during Early Life Has Persistent Effects on Natural Killer T Cell Function. Science 2012, 336, 489–493. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Hugot, J.P. CARD15/NOD2 Mutations in Crohn’s Disease. Ann. N. Y. Acad. Sci. 2006, 1072, 9–18. [Google Scholar] [CrossRef] [PubMed]
  75. Biank, V.; Broeckel, U.; Kugathasan, S. Pediatric Inflammatory Bowel Disease: Clinical and Molecular Genetics. Inflamm. Bowel Dis. 2007, 13, 1430–1438. [Google Scholar] [CrossRef] [PubMed]
  76. Salem, M.; Seidelin, J.B.; Rogler, G.; Nielsen, O.H. Muramyl Dipeptide Responsive Pathways in Crohn’s Disease: From NOD2 and Beyond. Cell. Mol. Life Sci. 2013, 70, 3391–3404. [Google Scholar] [CrossRef] [Green Version]
  77. Fritz, T.; Niederreiter, L.; Adolph, T.; Blumberg, R.S.; Kaser, A. Crohn’s Disease: NOD2, Autophagy and ER Stress Converge. Gut 2011, 60, 1580–1588. [Google Scholar] [CrossRef] [Green Version]
  78. Wehkamp, J.; Harder, J.; Weichenthal, M.; Schwab, M.; Schäffeler, E.; Schlee, M.; Herrlinger, K.R.; Stallmach, A.; Noack, F.; Fritz, P.; et al. NOD2 (CARD15) Mutations in Crohn’s Disease Are Associated with Diminished Mucosal Alpha-Defensin Expression. Gut 2004, 53, 1658–1664. [Google Scholar] [CrossRef] [Green Version]
  79. Kinose, D.; Ogawa, E.; Hirota, T.; Ito, I.; Kudo, M.; Haruna, A.; Marumo, S.; Hoshino, Y.; Muro, S.; Hirai, T.; et al. A NOD2 Gene Polymorphism Is Associated with the Prevalence and Severity of Chronic Obstructive Pulmonary Disease in a Japanese Population. Respirology 2012, 17, 164–171. [Google Scholar] [CrossRef]
  80. Scharl, M.; Rogler, G. Inflammatory Bowel Disease: Dysfunction of Autophagy? Dig. Dis. 2012, 30, 12–19. [Google Scholar] [CrossRef] [Green Version]
  81. Orchard, T.R.; Chua, C.N.; Ahmad, T.; Cheng, H.; Welsh, K.I.; Jewell, D.P. Uveitis and Erythema Nodosum in Inflammatory Bowel Disease: Clinical Features and the Role of HLA Genes. Gastroenterology 2002, 123, 714–718. [Google Scholar] [CrossRef] [PubMed]
  82. Roussomoustakaki, M.; Satsangi, J.; Welsh, K.; Louis, E.; Fanning, G.; Targan, S.; Landers, C.; Jewell, D.P. Genetic Markers May Predict Disease Behavior in Patients with Ulcerative Colitis. Gastroenterology 1997, 112, 1845–1853. [Google Scholar] [CrossRef]
  83. D’Andrea, N.; Vigliarolo, R.; Sanguinetti, C.M. Respiratory Involvement in Inflammatory Bowel Diseases. Multidiscip. Respir. Med. 2010, 5, 173–182. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Betancourt, S.L.; Palacio, D.; Jimenez, C.A.; Martinez, S.; Marom, E.M. Thoracic Manifestations of Inflammatory Bowel Disease. Am. J. Roentgenol. 2011, 197, W452–W456. [Google Scholar] [CrossRef] [PubMed]
  85. Lu, D.G.; Ji, X.Q.; Liu, X.; Li, H.J.; Zhang, C.Q. Pulmonary Manifestations of Crohn’s Disease. World J. Gastroenterol. 2014, 20, 133–141. [Google Scholar] [CrossRef]
  86. Russi, A.; Gurbani, N.; Rosen, M.J.; Mallon, D.; LeBlanc, F.R. Pediatric Patient with Ulcerative Colitis-Associated Bronchiectasis. ACG Case Rep. J. 2020, 7, e00365. [Google Scholar] [CrossRef]
  87. Camus, P.; Piard, F.; Ashcroft, T.; Gal, A.A.; Colby, T.V. The Lung in Inflammatory Bowel Disease. Medicine 1993, 72, 151–183. [Google Scholar] [CrossRef]
  88. Spira, A.; Grossman, R.; Balter, M. Large Airway Disease Associated with Inflammatory Bowel Disease. Chest 1998, 113, 1723–1726. [Google Scholar] [CrossRef]
  89. Maglione, M.; Aksamit, T.; Santamaria, F. Paediatric and Adult Bronchiectasis: Specific Management with Coexisting Asthma, COPD, Rheumatological Disease and Inflammatory Bowel Disease. Respirology 2019, 24, 1063–1072. [Google Scholar] [CrossRef]
  90. Mahadeva, R.; Walsh, G.; Flower, C.D.R.; Shneerson, J.M. Clinical and Radiological Characteristics of Lung Disease in Inflammatory Bowel Disease. Eur. Respir. J. 2000, 15, 41–48. [Google Scholar] [CrossRef]
  91. Songür, N.; Songür, Y.; Tüzün, M.; Doǧan, I.; Tüzün, D.; Ensari, A.; Hekimoglu, B. Pulmonary Function Tests and High-Resolution CT in the Detection of Pulmonary Involvement in Inflammatory Bowel Disease. J. Clin. Gastroenterol. 2003, 37, 292–298. [Google Scholar] [CrossRef] [PubMed]
  92. Douglas, J.G.; McDonald, C.F.; Leslie, M.J.; Gillon, J.; Crompton, G.K.; McHardy, G.J.R. Respiratory Impairment in Inflammatory Bowel Disease: Does It Vary with Disease Activity? Respir. Med. 1989, 83, 389–394. [Google Scholar] [CrossRef]
  93. Ceyhan, B.B.; Karakurt, S.; Cevik, H.; Sungur, M. Bronchial Hyperreactivity and Allergic Status in Inflammatory Bowel Disease. Respiration 2003, 70, 60–66. [Google Scholar] [CrossRef] [PubMed]
  94. Sato, H.; Okada, F.; Matsumoto, S.; Sonoda, A.; Murakami, K.; Ishida, T.; Takaki, H.; Wakisaka, M.; Tokuyama, K.; Shimada, R.; et al. Chest High-Resolution Computed Tomography Findings in 601 Patients with Inflammatory Bowel Diseases. Acad. Radiol. 2018, 25, 407–414. [Google Scholar] [CrossRef] [PubMed]
  95. Raymond, S.B.; Gee, M.S.; Anupindi, S.A.; Shailam, R.; Kaplan, J.L.; Nimkin, K. CT and MRI of Rare Extraintestinal Manifestations of Inflammatory Bowel Disease in Children and Adolescents. J. Pediatr. Gastroenterol. Nutr. 2016, 63, e1–e9. [Google Scholar] [CrossRef]
  96. Black, H.; Mendoza, M.; Murin, S. Thoracic Manifestations of Inflammatory Bowel Disease. Chest 2007, 131, 524–532. [Google Scholar] [CrossRef]
  97. Peradzyńska, J.; Krenke, K.; Lange, J.; Banaszkiewicz, A.; Łazowska-Przeorek, I.; Radzikowski, A.; Kulus, M. Low Prevalence of Pulmonary Involvement in Children with Inflammatory Bowel Disease. Respir. Med. 2012, 106, 1048–1054. [Google Scholar] [CrossRef] [Green Version]
  98. Furlano, R.I.; Basek, P.; Müller, P.; Bieli, C.; Braegger, C.P.; Barben, J.; Hammer, J.; Moeller, A.; Trachsel, D. Pulmonary Function Test Abnormalities in Pediatric Inflammatory Bowel Disease. Respiration 2015, 90, 279–286. [Google Scholar] [CrossRef]
  99. Ahmed, K.A.; Thompson, J.W.; Joyner, R.E.; Stocks, R.M.S. Airway Obstruction Secondary to Tracheobronchial Involvement of Asymptomatic Undiagnosed Crohn’s Disease in a Pediatric Patient. Int. J. Pediatr. Otorhinolaryngol. 2005, 69, 1003–1005. [Google Scholar] [CrossRef]
  100. Akobeng, A.K.; Miller, V.; Thomas, A.G. Unexplained Chronic Bronchial Suppuration and Inflammatory Bowel Disease. J. Pediatr. Gastroenterol. Nutr. 1999, 28, 324–326. [Google Scholar] [CrossRef]
  101. Bentur, L.; Lachter, J.; Koren, I.; Ben-Izhak, O.; Levy, A.; Bentur, Y.; Rosenthal, E. Severe Pulmonary Disease in Association with Crohn’s Disease in a 13-Year-Old Girl. Pediatr. Pulmonol. 2000, 29, 151–154. [Google Scholar] [CrossRef]
  102. Calder, C.J.; Lacy, D.; Raafat, F.; Weller, P.H.; Booth, I.W. Crohn’s Disease with Pulmonary Involvement in a 3 Year Old Boy. Gut 1993, 34, 1636–1638. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Chiaro, A.; Fries, W.; Romano, C. Pediatric Pulmonary Crohn’s Disease: More Frequent than Expected. J. Crohns Colitis 2013, 7, e189-90. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Kayser, K.; Probst, F.; Gabius, H.J.; Müller, K.M. Are There Characteristic Alterations of Lung Tissue Associated with Crohn’s Disease? Pathol. Res. Pract. 1990, 186, 485–490. [Google Scholar] [CrossRef]
  105. Krishnan, S.; Banquet, A.; Newman, L.; Katta, U.; Patil, A.; Dozor, A.J. Lung Lesions in Children with Crohn’s Disease Presenting as Nonresolving Pneumonias and Response to Infliximab Therapy. Pediatrics 2006, 117, 1440–1443. [Google Scholar] [CrossRef]
  106. Levenbrown, Y.; Tauber, D.; Hall, O.R.; Baldridge, A.D. Granulomatous Lung Disease as the Initial Presentation of Crohn Disease. J. Pediatr. Gastroenterol. Nutr. 2009, 48, 487–490. [Google Scholar] [CrossRef]
  107. Mahgoub, L.E.O.; Puntis, J.W.L.; Chetcuti, P.A.J.; Sugarman, I.D. Severe Crohn Disease of the Lung Following Colectomy. J. Pediatr. Gastroenterol. Nutr. 2007, 45, 477–479. [Google Scholar] [CrossRef]
  108. Minic, P.; Perisic, V.N.; Minic, A. Metastatic Crohn’s Disease of the Lung. J. Pediatr. Gastroenterol. Nutr. 1998, 27, 338–341. [Google Scholar] [CrossRef]
  109. Nelson, B.A.; Kaplan, J.L.; El Saleeby, C.M.; Lu, M.T.; Mark, E.J. Case Records of the Massachusetts General Hospital. Case 39-2014. A 9-Year-Old Girl with Crohn’s Disease and Pulmonary Nodules. N. Engl. J. Med. 2014, 371, 2418–2427. [Google Scholar] [CrossRef]
  110. Pain-Prado, E.; Rais, A.; Madhi, F.; Orzechowski, C.; Dubern, B.; Epaud, R. Pulmonary and Hepatic Nodular Lesions Precede the Diagnosis of Crohn’s Disease in an 8-Year-Old Girl: A Case Study and Review of the Literature. Acta Paediatr. Int. J. Paediatr. 2012, 101, e86–e89. [Google Scholar] [CrossRef]
  111. Puntis, J.W.L.; Tarlow, M.J.; Raafat, F.; Booth, I.W. Crohn’s Disease of the Lung. Arch. Dis. Child. 1990, 65, 1270–1271. [Google Scholar] [CrossRef] [PubMed]
  112. Mohsin Shah, S.; Clinton Texter, E.; White, H.J. Inflammatory Bowel Disease Associated with Granulomatous Lung Disease: Report of a Case with Endoscopic Findings. Gastrointest. Endosc. 1976, 23, 98–99. [Google Scholar] [CrossRef]
  113. Silbermintz, A.; Krishnan, S.; Banquet, A.; Markowitz, J. Granulomatous Pneumonitis, Sclerosing Cholangitis, and Pancreatitis in a Child with Crohn Disease: Response to Infliximab. J. Pediatr. Gastroenterol. Nutr. 2006, 42, 324–326. [Google Scholar] [PubMed]
  114. Valletta, E.; Bertini, M.; Sette, L.; Braggion, C.; Pradal, U.; Zannoni, M. Early Bronchopulmonary Involvement in Crohn Disease: A Case Report. BMC Gastroenterol. 2001, 1, 13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Basseri, B.; Enayati, P.; Marchevsky, A.; Papadakis, K.A. Pulmonary Manifestations of Inflammatory Bowel Disease: Case Presentations and Review. J. Crohn’s Colitis 2010, 4, 390–397. [Google Scholar] [CrossRef] [Green Version]
  116. Carvalho, R.S.; Wilson, L.; Cuffari, C. Pulmonary Manifestations in a Pediatric Patient with Ulcerative Colitis: A Case Report. J. Med. Case Rep. 2008, 2, 59. [Google Scholar] [CrossRef] [Green Version]
  117. Mazer, B.D.; Eigen, H.; Gelfand, E.W.; Brugman, S.M. Remission of Interstitial Lung Disease Following Therapy of Associated Ulcerative Colitis. Pediatr. Pulmonol. 1993, 15, 55–59. [Google Scholar] [CrossRef]
  118. Teague, W.G.; Sutphen, J.L.; Fechner, R.E. Desquamative Interstitial Pneumonitis Complicating Inflammatory Bowel Disease of Childhood. J. Pediatr. Gastroenterol. Nutr. 1985, 4, 663–667. [Google Scholar] [CrossRef]
  119. Herrlinger, K.R.; Noftz, M.K.; Dalhoff, K.; Ludwig, D.; Stange, E.F.; Fellermann, K. Alterations in Pulmonary Function in Inflammatory Bowel Disease Are Frequent and Persist during Remission. Am. J. Gastroenterol. 2002, 97, 377–381. [Google Scholar] [CrossRef]
  120. Tzanakis, N.; Samiou, M.; Bouros, D.; Mouzas, J.; Kouroumalis, E.; Siafakas, N.M. Small Airways Function in Patients with Inflammatory Bowel Disease. Am. J. Respir. Crit. Care Med. 1998, 157, 382–386. [Google Scholar] [CrossRef]
  121. Goyal, A.; Ghoshal, U.C.; Nath, A.; Jindal, S.; Mohindra, S. Pulmonary Function in Patients with Ulcerative Colitis and Its Relationship with Disease Severity. J. Gastroenterol. Hepatol. 2017, 1, 32–37. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Gupta, S.J.; Gupta, V.L.; Kothari, H.G.; Samarth, A.R.; Gaikwad, N.R.; Parmar, S.M. Assessment of Occult Pulmonary Involvement in Ulcerative Colitis. Inflamm. Intest. Dis. 2020, 5, 144–150. [Google Scholar] [CrossRef] [PubMed]
  123. Mohamed-Hussein, A.A.R.; Mohamed, N.A.S.; Ibrahim, M.E.A.R. Changes in Pulmonary Function in Patients with Ulcerative Colitis. Respir. Med. 2007, 101, 977–982. [Google Scholar] [CrossRef] [Green Version]
  124. Zhao, Y.; Wang, J.; Liu, Z.; Lin, H.; Shi, Y.; Sun, X. Pulmonary Dysfunction in 114 Patients with Inflammatory Bowel Disease. Medicine 2017, 96, e6808. [Google Scholar] [CrossRef] [PubMed]
  125. Welsh, L.; Haller, W.; King, L.E.; Soto-Martinez, M.; Oliver, M.; Catto-Smith, A.; Robinson, P.J. Pulmonary Function Abnormalities in Children with Active Crohn’s Disease. Am. J. Respir. Crit. Care Med. 2012, 186, 1060–1061. [Google Scholar] [CrossRef]
  126. Yammine, S.; Nyilas, S.; Casaulta, C.; Schibli, S.; Latzin, P.; Sokollik, C. Function and Ventilation of Large and Small Airways in Children and Adolescents with Inflammatory Bowel Disease. Inflamm. Bowel Dis. 2016, 22, 1915–1922. [Google Scholar] [CrossRef] [Green Version]
  127. Krenke, K.; Peradzyńska, J.; Lange, J.; Banaszkiewicz, A.; łazowska-Przeorek, I.; Grzela, K.; Radzikowski, A.; Kulus, M. Inflammatory Cytokines in Exhaled Breath Condensate in Children with Inflammatory Bowel Diseases. Pediatr. Pulmonol. 2014, 49, 1190–1195. [Google Scholar] [CrossRef]
  128. El Amrousy, D.M.; Hassan, S.; El-Ashry, H.; Yousef, M.; Sharshar, R. Pulmonary Function Tests Abnormalities in Children With Inflammatory Bowel Disease: Is It Common? J. Pediatr. Gastroenterol. Nutr. 2018, 67, 346–350. [Google Scholar] [CrossRef]
  129. Verstraete, M.; Choukroun, M.L.; Siao-Him Fa, V.; Fayon, M.; Rebouissoux, L.; Enaud, R.; Lamireau, T. Altered Pulmonary Gas Transfer Capacity and Capillary Blood Volume in Pediatric Crohn’s Disease. Pediatr. Pulmonol. 2017, 52, 1051–1056. [Google Scholar] [CrossRef]
  130. Munck, A.; Murciano, D.; Pariente, R.; Cezard, J.P.; Navarro, J. Latent Pulmonary Function Abnormalities in Children with Crohn’s Disease. Eur. Respir. J. 1995, 8, 377–380. [Google Scholar] [CrossRef]
  131. Barnes, P.J.; Dweik, R.A.; Gelb, A.F.; Gibson, P.G.; George, S.C.; Grasemann, H.; Pavord, I.D.; Ratjen, F.; Silkoff, P.E.; Taylor, D.R.; et al. Exhaled Nitric Oxide in Pulmonary Diseases: A Comprehensive Review. Chest 2010, 138, 682–692. [Google Scholar] [CrossRef] [PubMed]
  132. Martín, M.C.; Martinez, A.; Mendoza, J.L.; Taxonera, C.; Díaz-Rubio, M.; Fernández-Arquero, M.; De La Concha, E.G.; Urcelay, E. Influence of the Inducible Nitric Oxide Synthase Gene (NOS2A) on Inflammatory Bowel Disease Susceptibility. Immunogenetics 2007, 59, 833–837. [Google Scholar] [CrossRef] [PubMed]
  133. Lundberg, J.O.N.; Lundberg, J.M.; Alving, K.; Hellström, P.M. Greatly Increased Luminal Nitric Oxide in Ulcerative Colitis. Lancet 1994, 344, 1673–1674. [Google Scholar] [CrossRef]
  134. Gut, G.; Ben-Tov, A.; Lahad, A.; Soferman, R.; Cohen, S.; Tauman, R.; Sivan, Y. Pulmonary Functions in Children with Inflammatory Bowel Disease. Eur. J. Gastroenterol. Hepatol. 2016, 28, 708–713. [Google Scholar] [CrossRef] [PubMed]
  135. Livnat, G.; Lerner, A.; Hakim, F.; Eshach-Adiv, O.; Berkowitz, D.; Bentur, L. Bronchial Reactivity and Fractional Exhaled NO in Crohn’s Disease in the Era of Immunomodulating Treatment. Acta Paediatr. 2012, 101, e399–e404. [Google Scholar] [CrossRef] [PubMed]
  136. Huang, Y.; Lemberg, D.A.; Day, A.S.; Dixon, B.; LeaC.H., S.; Bujanover, Y.; Jaffe, A.; Thomas, P.S. Markers of Inflammation in the Breath in Paediatric Inflammatory Bowel Disease. J. Pediatr. Gastroenterol. Nutr. 2014, 59, 505–510. [Google Scholar] [CrossRef]
  137. Koek, G.H.; Verleden, G.M.; Evenepoel, P.; Rutgeerts, P. Activity Related Increase of Exhaled Nitric Oxide in Crohn’s Disease and Ulcerative Colitis: A Manifestation of Systemic Involvement? Respir. Med. 2002, 96, 530–535. [Google Scholar] [CrossRef] [Green Version]
  138. Malerba, M.; Ragnoli, B.; Buffoli, L.; Radaeli, A.; Ricci, C.; Lanzarotto, F.; Lanzini, A. Exhaled Nitric Oxide as a Marker of Lung Involvement in Crohn’s Disease. Int. J. Immunopathol. Pharmacol. 2011, 24, 1119–1124. [Google Scholar] [CrossRef]
  139. Shoman, Y.; Wild, P.; Hemmendinger, M.; Graille, M.; Sauvain, J.J.; Hopf, N.B.; Canu, I.G. Reference Ranges of 8-Isoprostane Concentrations in Exhaled Breath Condensate (EBC): A Systematic Review and Meta-Analysis. Int. J. Mol. Sci. 2020, 21, 3822. [Google Scholar] [CrossRef]
  140. Hensel, K.O.; Boland, V.; Postberg, J.; Zilbauer, M.; Heuschkel, R.; Vogel, S.; Gödde, D.; Wirth, S.; Jenke, A.C. Differential Expression of Mucosal Trefoil Factors and Mucins in Pediatric Inflammatory Bowel Diseases. Sci. Rep. 2014, 4, 7343. [Google Scholar] [CrossRef] [Green Version]
  141. Mansi, A.; Cucchiara, S.; Greco, L.; Sarnelli, P.; Pisanti, C.; Franco, M.T.; Santamaria, F. Bronchial Hyperresponsiveness in Children and Adolescents with Crohn’s Disease. Am. J. Respir. Crit. Care Med. 2000, 161, 1051–1054. [Google Scholar] [CrossRef] [PubMed]
  142. Massart, A.; Hunt, D.P. Pulmonary Manifestations of Inflammatory Bowel Disease. Am. J. Med. 2020, 133, 39–43. [Google Scholar] [CrossRef] [PubMed]
  143. Van Rheenen, P.F.; Aloi, M.; Assa, A.; Bronsky, J.; Escher, J.C.; Fagerberg, U.L.; Gasparetto, M.; Gerasimidis, K.; Griffiths, A.; Henderson, P.; et al. The Medical Management of Paediatric Crohn’s Disease: An ECCO-ESPGHAN Guideline Update. J. Crohns Colitis 2020, 15, 171–194. [Google Scholar] [CrossRef] [PubMed]
  144. Turner, D.; Ruemmele, F.M.; Orlanski-Meyer, E.; Griffiths, A.M.; De Carpi, J.M.; Bronsky, J.; Veres, G.; Aloi, M.; Strisciuglio, C.; Braegger, C.P.; et al. Management of Paediatric Ulcerative Colitis, Part 1: Ambulatory Care—An Evidence-Based Guideline From European Crohn’s and Colitis Organization and European Society of Paediatric Gastroenterology, Hepatology and Nutrition. J. Pediatr. Gastroenterol. Nutr. 2018, 67, 257–291. [Google Scholar] [CrossRef] [PubMed]
  145. Harbord, M.; Annese, V.; Vavricka, S.R.; Allez, M.; de Acosta, M.B.; Boberg, K.M.; Burisch, J.; Martine, D.V.; De Vries, A.M.; Dick, A.D.; et al. The First European Evidence-Based Consensus on Extra-Intestinal Manifestations in Inflammatory Bowel Disease. J. Crohns Colitis 2016, 10, 239–254. [Google Scholar] [CrossRef]
  146. Foster, R.A.; Zander, D.S.; Mergo, P.J.; Valentine, J.F. Mesalamine-Related Lung Disease: Clinical, Radiographic, and Pathologic Manifestations. Inflamm. Bowel Dis. 2003, 9, 308–315. [Google Scholar] [CrossRef]
  147. Boyd, O.; Gibbs, A.R.; Smith, A.P. Fibrosing Alveolitis Due to Sulphasalazine in a Patient with Rheumatoid Arthritis. Br. J. Rheumatol. 1990, 29, 222–224. [Google Scholar] [CrossRef]
  148. Gupta, A.; Gulati, S. Mesalamine Induced Eosinophilic Pneumonia. Respir. Med. Case Rep. 2017, 21, 116–117. [Google Scholar] [CrossRef]
  149. Borchers, A.T.; Keen, C.L.; Cheema, G.S.; Gershwin, M.E. The Use of Methotrexate in Rheumatoid Arthritis. Semin. Arthritis Rheum. 2004, 34, 465–483. [Google Scholar] [CrossRef]
  150. Schnabel, A.; Gross, W.L. Low-Dose Methotrexate in Rheumatic Diseases--Efficacy, Side Effects, and Risk Factors for Side Effects. Semin. Arthritis Rheum. 1994, 23, 310–327. [Google Scholar] [CrossRef]
  151. Imokawa, S.; Colby, T.V.; Leslie, K.O.; Helmers, R.A. Methotrexate Pneumonitis: Review of the Literature and Histopathological Findings in Nine Patients. Eur. Respir. J. 2000, 15, 373–381. [Google Scholar] [CrossRef] [PubMed]
  152. Lee, H.S.; Jo, K.W.; Shim, T.S.; Song, J.W.; Lee, H.J.; Hwang, S.W.; Park, S.H.; Yang, D.H.; Kim, K.J.; Ye, B.D.; et al. Six Cases of Lung Injury Following Anti-Tumour Necrosis Factor Therapy for Inflammatory Bowel Disease. J. Crohns Colitis 2015, 9, 1053–1057. [Google Scholar] [CrossRef] [Green Version]
  153. Stevens, J.P.; Ballengee, C.R.; Chandradevan, R.; Thompson, A.B.; Schoen, B.T.; Kugathasan, S.; Sauer, C.G. Performance of Interferon-Gamma Release Assays for Tuberculosis Screening in Pediatric Inflammatory Bowel Disease. J. Pediatr. Gastroenterol. Nutr. 2019, 69, e111–e116. [Google Scholar] [CrossRef] [PubMed]
  154. Byun, J.M.; Lee, C.K.; Rhee, S.Y.; Kim, H.J.; Im, J.P.; Park, D., II; Eun, C.S.; Jung, S.A.; Shin, J.E.; Lee, K.M.; et al. Risks for Opportunistic Tuberculosis Infection in a Cohort of 873 Patients with Inflammatory Bowel Disease Receiving a Tumor Necrosis Factor-α Inhibitor. Scand. J. Gastroenterol. 2015, 50, 312–320. [Google Scholar] [CrossRef] [PubMed]
  155. Winthrop, K.L.; Chang, E.; Yamashita, S.; Iademarco, M.F.; LoBue, P.A. Nontuberculous Mycobacteria Infections and Anti–Tumor Necrosis Factor-α Therapy. Emerg. Infect. Dis. 2009, 15, 1556–1561. [Google Scholar] [CrossRef] [PubMed]
  156. Jansson-Knodell, C.L.; Harris, C.E.; Loftus, E.V.; Walker, R.C.; Enzler, M.J.; Virk, A. Histoplasmosis in Inflammatory Bowel Disease with Tumor Necrosis Factor-Alpha Inhibitors: Safe to Continue Biologics? Dig. Dis. Sci. 2021, 66, 190–198. [Google Scholar] [CrossRef]
  157. Warris, A.; Bjørneklett, A.; Gaustad, P. Invasive Pulmonary Aspergillosis Associated with Infliximab Therapy. N. Engl. J. Med. 2001, 344, 1099–1100. [Google Scholar]
  158. Hirai, F.; Matsui, T.; Ishibashi, Y.; Higashi, D.; Futami, K.; Haraoka, S.; Iwashita, A. Asymptomatic Pulmonary Cryptococcosis in a Patient with Crohn’s Disease on Infliximab: Case Report. Inflamm. Bowel Dis. 2011, 17, 1637–1638. [Google Scholar] [CrossRef]
  159. Lawrance, I.C.; Radford-Smith, G.L.; Bampton, P.A.; Andrews, J.M.; Tan, P.K.; Croft, A.; Gearry, R.B.; Florin, T.H.J. Serious Infections in Patients with Inflammatory Bowel Disease Receiving Anti-Tumor-Necrosis-Factor-Alpha Therapy: An Australian and New Zealand Experience. J. Gastroenterol. Hepatol. 2010, 25, 1732–1738. [Google Scholar] [CrossRef]
  160. Tschudy, J.; Michail, S. Disseminated Histoplasmosis and Pneumocystis Pneumonia in a Child with Crohn Disease Receiving Infliximab. J. Pediatr. Gastroenterol. Nutr. 2010, 51, 221–222. [Google Scholar] [CrossRef]
  161. Verma, R.; Walia, R.; Sondike, S.B.; Khan, R. Pulmonary Nocardiosis in an Adolescent Patient with Crohn’s Disease Treated with Infliximab: A Serious Complication of TNF-Alpha Blockers. W. Va. Med. J. 2015, 111, 36–39. [Google Scholar] [PubMed]
  162. Kaur, N.; Mahl, T.C. Pneumocystis Jiroveci (Carinii) Pneumonia after Infliximab Therapy: A Review of 84 Cases. Dig. Dis. Sci. 2007, 52, 1481–1484. [Google Scholar] [CrossRef] [PubMed]
  163. Tragiannidis, A.; Kyriakidis, I.; Zündorf, I.; Groll, A.H. Invasive Fungal Infections in Pediatric Patients Treated with Tumor Necrosis Alpha (TNF-α) Inhibitors. Mycoses 2017, 60, 222–229. [Google Scholar] [CrossRef]
  164. Toussi, S.S.; Pan, N.; Walters, H.M.; Walsh, T.J. Infections in Children and Adolescents with Juvenile Idiopathic Arthritis and Inflammatory Bowel Disease Treated with Tumor Necrosis Factor-α Inhibitors: Systematic Review of the Literature. Clin. Infect. Dis. 2013, 57, 1318–1330. [Google Scholar] [CrossRef]
  165. Villeneuve, E.; St-Pierre, A.; Haraoui, B. Interstitial Pneumonitis Associated with Infliximab Therapy. J. Rheumatol. 2006, 33, 1189–1193. [Google Scholar] [PubMed]
  166. Karampitsakos, T.; Papaioannou, O.; Sampsonas, F.; Tzouvelekis, A. Infliximab-Induced Interstitial Lung Disease. BMJ Case Rep. 2021, 14, e245726. [Google Scholar] [CrossRef]
  167. Veerappan, S.G.; O’Morain, C.A. Infliximab-Associated Alveolitis after Treatment for Severe Left-Sided Ulcerative Colitis. Eur. J. Gastroenterol. Hepatol. 2009, 21, 830–832. [Google Scholar] [CrossRef]
  168. Panagi, S.; Palka, W.; Korelitz, B.I.; Taskin, M.; Lessnau, K.D. Diffuse Alveolar Hemorrhage after Infliximab Treatment of Crohn’s Disease. Inflamm. Bowel Dis. 2004, 10, 274–277. [Google Scholar] [CrossRef]
  169. Sen, S.; Peltz, C.; Jordan, K.; Boes, T.J. Infliximab-Induced Nonspecific Interstitial Pneumonia. Am. J. Med. Sci. 2012, 344, 75–78. [Google Scholar] [CrossRef]
  170. Cucinotta, U.; Dipasquale, V.; Costa, S.; Pellegrino, S.; Ramistella, V.; Romano, C. Vedolizumab-Associated Pulmonary Manifestations in Children with Ulcerative Colitis. J. Clin. Pharm. Ther. 2022, 47, 254–256. [Google Scholar] [CrossRef]
  171. Mitchel, E.B.; Paul, A.; El-Ali, A.; Cheng, P.C.; Albenberg, L.G. Drug-Induced Lung Disease Associated With Ustekinumab in a Pediatric Patient With Crohn Disease. J. Pediatr. Gastroenterol. Nutr. 2020, 71, e143–e145. [Google Scholar] [CrossRef] [PubMed]
  172. Certain, M.C.; Georges, M.; Beltramo, G.; Schenesse, D.; Mouillot, P.; Tankéré, P.; Bonniaud, P. Adalimumab-Induced Interstitial Pneumonia in Patients with Inflammatory Bowel Disease. Respir. Med. Res. 2020, 78, 100790. [Google Scholar] [CrossRef] [PubMed]
  173. Imdad, A.; Nicholson, M.R.; Tanner-Smith, E.E.; Zackular, J.P.; Gomez-Duarte, O.G.; Beaulieu, D.B.; Acra, S. Fecal Transplantation for Treatment of Inflammatory Bowel Disease. Cochrane Database Syst. Rev. 2018, 11, CD012774. [Google Scholar] [CrossRef] [PubMed]
  174. Fehily, S.R.; Basnayake, C.; Wright, E.K.; Kamm, M.A. Fecal Microbiota Transplantation Therapy in Crohn’s Disease: Systematic Review. J. Gastroenterol. Hepatol. 2021, 36, 2672–2686. [Google Scholar] [CrossRef]
  175. Costello, S.; Waters, O.; Bryant, R.; Katsikeros, R.; Makanyanga, J.; Schoeman, M.N.; Mountifield, R.E.; Tee, D.; Howell, S.C.; Hughes, P.A.; et al. Short Duration, Low Intensity Pooled Faecal Microbiota Transplantation Induces Remission in Patients with Mild-Moderately Active Ulcerative Colitis: A Randomised Controlled Trial. J. Crohn’s Colitis 2017, 11, S23. [Google Scholar] [CrossRef] [Green Version]
  176. Lazzerini, M.; Bramuzzo, M.; Maschio, M.; Martelossi, S.; Ventura, A. Thromboembolism in Pediatric Inflammatory Bowel Disease: Systematic Review. Inflamm. Bowel Dis. 2011, 17, 2174–2183. [Google Scholar] [CrossRef]
  177. Zitomersky, N.L.; Levine, A.E.; Atkinson, B.J.; Harney, K.M.; Verhave, M.; Bousvaros, A.; Lightdale, J.R.; Trenor, C.C. Risk Factors, Morbidity, and Treatment of Thrombosis in Children and Young Adults with Active Inflammatory Bowel Disease. J. Pediatr. Gastroenterol. Nutr. 2013, 57, 343–347. [Google Scholar] [CrossRef]
  178. Kuenzig, M.E.; Bitton, A.; Carroll, M.W.; Kaplan, G.G.; Otley, A.R.; Singh, H.; Nguyen, G.C.; Griffiths, A.M.; Stukel, T.A.; Targownik, L.E.; et al. Inflammatory Bowel Disease Increases the Risk of Venous Thromboembolism in Children: A Population-Based Matched Cohort Study. J. Crohns Colitis 2021, 15, 2031–2040. [Google Scholar] [CrossRef]
  179. Kappelman, M.D.; Horvath-Puho, E.; Sandler, R.S.; Rubin, D.T.; Ullman, T.A.; Pedersen, L.; Baron, J.A.; Sørensen, H.T. Thromboembolic Risk among Danish Children and Adults with Inflammatory Bowel Diseases: A Population-Based Nationwide Study. Gut 2011, 60, 937–943. [Google Scholar] [CrossRef]
  180. Nylund, C.M.; Goudie, A.; Garza, J.M.; Crouch, G.; Denson, L.A. Venous Thrombotic Events in Hospitalized Children and Adolescents with Inflammatory Bowel Disease. J. Pediatr. Gastroenterol. Nutr. 2013, 56, 485–491. [Google Scholar] [CrossRef]
  181. Giannotta, M.; Tapete, G.; Emmi, G.; Silvestri, E.; Milla, M. Thrombosis in Inflammatory Bowel Diseases: What’s the Link? Thromb. J. 2015, 13, 14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  182. Singh, U.; Devaraj, S.; Jialal, I. C-Reactive Protein Decreases Tissue Plasminogen Activator Activity in Human Aortic Endothelial Cells: Evidence That C-Reactive Protein Is a Procoagulant. Arterioscler. Thromb. Vasc. Biol. 2005, 25, 2216–2221. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  183. Chen, C.; Nan, B.; Lin, P.; Yao, Q. C-Reactive Protein Increases Plasminogen Activator Inhibitor-1 Expression in Human Endothelial Cells. Thromb. Res. 2008, 122, 125–133. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  184. Van Der Poll, T.; Jansen, P.M.; Van Zee, K.J.; Burress Welborn, M.; De Jong, I.; Hack, C.E.; Loetscher, H.; Lesslauer, W.; Lowry, S.F.; Moldawer, L.L. Tumor Necrosis Factor-Alpha Induces Activation of Coagulation and Fibrinolysis in Baboons through an Exclusive Effect on the P55 Receptor. Blood 1996, 88, 922–927. [Google Scholar] [CrossRef]
  185. Stouthard, J.M.L.; Levi, M.; Hack, C.E.; Veenhof, C.H.N.; Romijn, H.A.; Sauerwein, H.P.; Van Der Poll, T. Interleukin-6 Stimulates Coagulation, Not Fibrinolysis, in Humans. Thromb. Haemost. 1996, 76, 738–742. [Google Scholar] [CrossRef]
  186. Turner, D.; Ruemmele, F.M.; Orlanski-Meyer, E.; Griffiths, A.M.; De Carpi, J.M.; Bronsky, J.; Veres, G.; Aloi, M.; Strisciuglio, C.; Braegger, C.P.; et al. Management of Paediatric Ulcerative Colitis, Part 2: Acute Severe Colitis—An Evidence-Based Consensus Guideline From the European Crohn’s and Colitis Organization and the European Society of Paediatric Gastroenterology, Hepatology and Nutrition. J. Pediatr. Gastroenterol. Nutr. 2018, 67, 292–310. [Google Scholar] [CrossRef]
  187. Aardoom, M.A.; Klomberg, R.C.W.; Kemos, P.; Ruemmele, F.M.; van Ommen, C.H.; de Ridder, L.; Croft, N.M.; Turner, D.; Focht, G.; Croft, N.; et al. The Incidence and Characteristics of Venous Thromboembolisms in Paediatric-Onset Inflammatory Bowel Disease; a Prospective International Cohort Study Based on the PIBD-SETQuality Safety Registry. J. Crohns Colitis 2021, 16, 695–707. [Google Scholar] [CrossRef]
  188. Jochmann, A.; Trachsel, D.; Hammer, J. Inflammatory Bowel Disease and the Lung in Paediatric Patients. Breathe 2021, 17, 200269. [Google Scholar] [CrossRef]
  189. Majewski, S.; Piotrowski, W. Pulmonary manifestations of inflammatory bowel disease. Arch. Med. Sci. 2015, 11, 1179–1188. [Google Scholar] [CrossRef]
  190. Hayek, A.J.; Pfanner, T.P.; White, H.D. Inflammatory Bowel Disease of the Lung: The Role of Infliximab? Respir. Med. Case Rep. 2015, 15, 85–88. [Google Scholar] [CrossRef] [Green Version]
  191. Jain, R.; Hachem, R.R.; Morrell, M.R.; Trulock, E.P.; Chakinala, M.M.; Yusen, R.D.; Huang, H.J.; Mohanakumar, T.; Patterson, G.A.; Walter, M.J. Azithromycin Is Associated with Increased Survival in Lung Transplant Recipients with Bronchiolitis Obliterans Syndrome. J. Heart Lung Transplant. 2010, 29, 531–537. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  192. Camus, P.; Colby, T.V. The Lung in Inflammatory Bowel Disease. Eur. Respir. J. 2000, 15, 5–10. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Different pathophysiological concepts of pulmonary manifestations in IBD patients. (1) Mutation of the CARD-15 gene leads to a diminished expression of α-defensins and consequently to a breakdown of the mucosal barrier. (2) Less Clostridia spp. result in dysregulation of host–commensal relationship and, via the reduction of IL-22, to the alteration of epithelial junction integrity. (3) Luminal antigens penetrate through the leaky intestinal mucosa and activate dendritic cells (DC). (4) Activated macrophages (ΜΦ) lead, via IL-1 and TNF-α, to the expression of neutrophils (Nc) adhesion molecules. (5) Nc undergo margination and diapedesis to the intestinal mucosa. (6) Nc become primed and a subset re-migrates to the bloodstream. (7) Nc change their shape and become deformable, resulting in a prolonged transit time in pulmonary capillaries. (8) JAM-C becomes downregulated and therefore the translocation of Nc to respiratory epithelium is facilitated. (9) The injured lung is not able to de-prime Nc leading to more neutrophilic inflammation. (10) Inflamed intestinal tissue leads to the upregulation of non-tissue-specific T-cell receptors CCR3 and CXCR5. (11) Diapedesis of gut-memory T cells (Tc)) via non-specific endothelial receptors to lung tissue is increased. (12) Collagen becomes degraded to tripeptide proline-glycine-proline, which plays a role in chemotaxis of Nc. (13) Increased bacterial burden leads to the expression of platelet-activating factor receptor and subsequently to the overexpression of the inflammasome. (14) Gut and lung tissues share the same embryological origin in the foregut portion of the endoderm. Abbreviations: iEC, intestinal epithelial cell; rEC, respiratory epithelial cell; Gc, goblet cell; Pc, Paneth cell; T1c, Type 1 pneumocytes; T2c, Type 2 pneumocytes; IL-1, interleukin 1; TNF-α, tumor necrosis factor alpha; pNC, primed neutrophils.
Figure 1. Different pathophysiological concepts of pulmonary manifestations in IBD patients. (1) Mutation of the CARD-15 gene leads to a diminished expression of α-defensins and consequently to a breakdown of the mucosal barrier. (2) Less Clostridia spp. result in dysregulation of host–commensal relationship and, via the reduction of IL-22, to the alteration of epithelial junction integrity. (3) Luminal antigens penetrate through the leaky intestinal mucosa and activate dendritic cells (DC). (4) Activated macrophages (ΜΦ) lead, via IL-1 and TNF-α, to the expression of neutrophils (Nc) adhesion molecules. (5) Nc undergo margination and diapedesis to the intestinal mucosa. (6) Nc become primed and a subset re-migrates to the bloodstream. (7) Nc change their shape and become deformable, resulting in a prolonged transit time in pulmonary capillaries. (8) JAM-C becomes downregulated and therefore the translocation of Nc to respiratory epithelium is facilitated. (9) The injured lung is not able to de-prime Nc leading to more neutrophilic inflammation. (10) Inflamed intestinal tissue leads to the upregulation of non-tissue-specific T-cell receptors CCR3 and CXCR5. (11) Diapedesis of gut-memory T cells (Tc)) via non-specific endothelial receptors to lung tissue is increased. (12) Collagen becomes degraded to tripeptide proline-glycine-proline, which plays a role in chemotaxis of Nc. (13) Increased bacterial burden leads to the expression of platelet-activating factor receptor and subsequently to the overexpression of the inflammasome. (14) Gut and lung tissues share the same embryological origin in the foregut portion of the endoderm. Abbreviations: iEC, intestinal epithelial cell; rEC, respiratory epithelial cell; Gc, goblet cell; Pc, Paneth cell; T1c, Type 1 pneumocytes; T2c, Type 2 pneumocytes; IL-1, interleukin 1; TNF-α, tumor necrosis factor alpha; pNC, primed neutrophils.
Ijms 23 07287 g001
Table 1. Published cases of pulmonary manifestation in pediatric CD patients.
Table 1. Published cases of pulmonary manifestation in pediatric CD patients.
First AuthorPublication YearAgeSexδPulmonary SymptomsRadiographic EvaluationPFTHistopathology (Lung)TherapyOutcome
Ahmed [99]20059yFSame timeStridor, shortness of breathXR: Normal
BSC: Diffuse hemorrhagic tracheitis
Not performedTracheal biopsy: granulomatous inflammationNot mentionedClinical improvement after 7mo of CD therapy
Akobeng [100]19999yM4y (b)Breathlessness, cough, wheezeXR: Unilateral consolidationNot performedNot performedBronchodilators, antibiotics, physiotherapyLittle initial effect.
No outcome mentioned after initiation of CD therapy
Al-Binali [11]200311yMSame
time
Exercise limitation, cough, shortness of breathXR: Patchy airspace consolidation
CT-T: Multiple peripheral nodules
AbnormalNon-caseating epithelioid granulomatous inflammationNot performedResolution of symptoms, radiological normalization and PFT improvement without therapy
Bentur [101]200013yF10moShortness of breathXR: Normal
CT-T: Patches of ground glass-appearing lung alternating with normal appearing areas of lung
AbnormalMononuclear inflammation with small non-caseating granulomas, bronchiolitis obliterans.1. PRD, 6-MP
2. Hydroxy-chloroquine
3. After severe bone marrow depression switch to PRD + MES
Slow improvement of symptoms and PFT with combination therapy of PRD + MES
Calder [102]19933yMSame
time
NoneXR: Unilateral densitiesNot performedNon-caseating epithelioid granulomatous inflammationNot mentionedNot mentioned
Chiaro [103]201314yF4yNon-productive cough, dyspnea, wheezing, chest painXR: Normal
CT-T: Multiple pulmonary nodules, parenchymal infiltrates, diffuse bilateral interstitial inflammation
Not performedNon-caseating granulomatous inflammation with multinucleated giant cellsOral PRDClinical and radiological improvement after 2mo
Inoue [14]201714yM14yShortness of breath, right pleuritic chest painXR + CT-T: Atelectasis, infiltration, pleural effusionNot performedPolypoid fibrosis, inflammatory cell infiltration1. Intravenous Pulse-MP
2. Oral PRD
Complete resolution of symptoms soon after initiation of MP
Kayser [104]199012yM3yTachypnea, dyspnea, increased mucus productionNot performedNot performedGranulomatous interstitial inflammation 1. Oral PRD
2. AZA
Improvement after initiation of AZA leading to improvement after 2y
Krishnan [105]200613yF4yBackpain, coughXR: “Round pneumonia”
CT-T: Multiple pleural and intraparenchymal infiltrates
AbnormalNon-caseating epithelioid granulomatous inflammation with giant cells1. Oral PRD
2. IFX
Immediate clinical and radiological response after initiation of IFX
Krishnan [105]200614yF3yChest tightness, shortness of breath, coughXR: Unilateral infiltrate
CT-T: Multiple, scattered granulomatous lesions
AbnormalNon-caseating epithelioid granulomatous inflammation with giant cellsIFXRapid clinical and radiological complete resolution
Levenbrown [106]200915yFSame
time
Non-productive coughXR: Bilateral patchy areas of opacification.
CT-T: Bilateral peribronchovascular nodules and patchy infiltration
AbnormalNon-caseating granulomatous inflammation with multinucleated giant cells, histiocytes, acute and chronic inflammation cell infiltrates. Organizing pneumonia.PRD, MTX, MESMild improvement of PFT 4d after initiation of therapy. No comment on clinical improvement.
Mahgoub [107]20075yM4yCough, shortness of breathXR: Extensive airspace shadowing, ring shadows.
CT-T: Interstitial lung disease
Not performedNon-caseating epithelioid granulomatous inflammation, lymphocytic interstitial pneumonitis1. Oral PRD
2. AZA
Only little overall improvement. Continuation of exercise-induced hypoxia. Lung transplantation was considered
Minic [108]199815yFSame timeExercise-induced dyspnea, coughXR: Central density of the right middle lobe with a minor interlobular effusionNormalSubepithelial, non-caseating epithelioid granulomatous inflammationOral PRD, SUFResolution of symptoms after 2mo
Nelson [109]20149yF2yCough, pleuritic chest pain, dyspnea on exertionCT-T: Multiple subpleural lung nodulesAbnormalGranulomatous inflammation, microabscesses with neutrophils and eosinophils1. Oral PRD
2. IFX
Resolution of symptoms after 1mo. Relapse after discontinuation of PRD. Initiation of IFX led to complete resolution.
Pain-Prado [110]20128yF5mo (b)TachypneaXR: Interstitial infiltration
CT-T: Multiple disseminated irregular stellar nodules
NormalNot performedNo treatmentComplete resolution without treatment
Puntis [111]199017yM2yUnilateral pleuritic chest pain, productive cough, dyspneaXR: Unilateral patchy consolidation, small pleural effusionNot performedNon-caseating epithelioid granulomatous inflammationIsoniazid, RifampicinComplete resolution after 6w
Shah [112]197613yM2y (b)CoughXR: Bilateral reticulonodular patternNot performedInterstitial pneumonitis with multiple non-caseating granulomas PRD, ferrous sulfateNot mentioned
Silbermintz [113]200613yF3yBackpain, coughXR: Bilateral infiltrates and densities
CT-T: 2 rounded densities, multiple pleural and intraparenchymal pulmonary lesions
AbnormalNon-caseating epithelioid granulomatous inflammation without interstitial fibrosis IFXFast resolution of symptoms and improvement and XR
Taylor [18]202010yM3yNoneCT-T: Bilateral pulmonary nodules.Not performedNon-necrotizing granulomatous bronchiolitisIncreased dose of ADAPulmonary nodules decreased
Taylor [18]202017yFSame
time
NoneCT-T: Bilateral “tree-in-bud” opacitiesNot performedNecrotizing granulomatous inflammation and focal organizing pneumoniaCorticosteroidsImprovement on CT-scans
Taylor [18]202014yM1moNon-productive coughCT-T: Diffuse pulmonary micronoduleNot performedGranulomatous inflammationNoneSymptom improvement shortly after starting maintenance therapy (IFX + MTX). Repeat CT-T 1y later with regression of pulmonary nodules
Taylor [18]202014yF1yNon-productive cough, pleuritic chest painXR: Right upper lobe nodule
CT-T: Bilateral pulmonary nodules
Not performedChronic interstitial pneumonitis with patchy organizing pneumonia without granulomasNoneSymptom improvement. CT-T 8mo later with resolution of previous nodules, appearance of 2 new nodules
Vadlamudi [17]201211yFSame
time
Non-productive coughXR: Unilateral consolidation and pleural effusionNot performedNot performedPRD, IFXClinical and radiological improvement
Vadlamudi [17]201217yF5yCough, shortness of breathXR: Bilateral opacities
CT-T: Bilateral cavitary lesions
Not performedNot performedIFXClinical and radiological improvement
Valletta [114]20016yF5moCoughXR: Unilateral parenchymal densityNormalThickening of basal membrane, angiectasis, active chronic inflammation1. Antibiotics
2. PRD
Prompt resolution of symptoms with antibiotics, then after 2mo relapse with cessation of symptoms after short-term therapy with PRD
Abbreviations: δ, time between diagnosis of inflammatory bowel disease (IBD) and pulmonary manifestation (PM); y, years; mo, months; w, weeks; d, days; M, male; F, female; PFT, pulmonary function tests; (b), diagnosis of PM preceding IBD; XR, chest x-ray; CT-T, thoracic computed tomography; BSC, bronchoscopy; PRD, prednisolone; M, methylprednisolone; IFX, infliximab; ADA, adalimumab; AZA, azathioprine; MES, mesalamine; MTX, methotrexate; 6-MP, 6-mercaptopurine; SUF, sulfasalazine.
Table 2. Published cases of pulmonary manifestations in pediatric UC patients.
Table 2. Published cases of pulmonary manifestations in pediatric UC patients.
First AuthorPublication YearAgeSexδPulmonary SymptomsRadiographic EvaluationPFTHistopathology (Lung)TherapyOutcome
Basseri [115]201017yM1yUnilateral chest pain, shortness of breathXR: Multiple bilateral patchy nodular opacities
CT-T: Multiple bilateral nodules of varying sizes
Not performedMultifocal acute alveolitis with multiple abscesses PRDClinical and radiological improvement after 1mo
Carvalho [116]200813yF1yNoneCT-T: 3 Nodules in lingula and left lower lobeAbnormalMarked alveolar inflammation, necrobiotic noduleIntravenous steroids, parental nutrition, antibiotics, MES, 6-MPResolution of nodules in follow-up CT-T 12mo later
Gut [25]201115yF1yProductive cough, wheezingXR: Mild bilateral hyperinflammation, bronchial thickening
CT-T: Diffuse bronchiectasis of all lung fields
AbnormalNot performedMacrolides, high-dose steroids (budesonide), bronchodilators, physical therapyProgressive improvement over 6mo
Krishnan [105]200617yM4yChest painXR: Bilateral basal infiltrates and pleural effusionNot performedBronchiolitis obliterans and organizing pneumonia1. Oral PRD
2. IFX
Rapid clinical and radiological complete resolution
Mazer [117]199313yF2y (b)Non-productive cough, shortness of breathXR: Bilateral diffuse interstitial infiltrate in a reticular pattern, peribronchial thickening, bronchiectasis, nodulesAbnormalNecrotizing bronchiolitis and bronchiectasis with interstitial pneumonitis1. Oral PRD
2. SUF
Improvement after initiation of SUF, relapse 8mo after discontinuation of PRD. Improvement after re-implementation of PRD
Russi [86]202017yF6yChronic purulent coughXR: Normal
CT-T: “Tree-in-but” opacities, mild bronchiectasis, bronchial wall thickening
AbnormalNot performedMP, albuterol and inhalation with 3% hypertonic salineClinical improvement. Relapse 1y later with another therapy cycle and following improvement
Taylor [18]202016yM1yPleuritic chest painXR: Bilateral pulmonary opacities
CT-T: Multiple pulmonary nodules
Not performed1. Biopsy: Necrotic neutrophilic nodules without granulomas1. PRD
2. IFX
Initial resolution of symptoms. Recurrence 1y later during IBD flare, with dramatic improvement after initial of IFX
Taylor [18]202013yF1yChronic productive cough, exertional dyspneaXR: Multiple pulmonary nodules
CT-T: Multiple pulmonary nodules, organizing pneumonia
AbnormalCryptogenic organizing pneumonia without granulomasFluticasone and albuterol inhalationCT-T 2mo later with improvement of nodules and no evidence of pneumonia. Improvement of cough, but continuous dyspnea on exertion
Teague [118]198512yM5yExertional dyspneaXR: Extensive, bilateral nodular interstitial patternAbnormalDesquamative interstitial pneumonitis1. Oral PRD
2. AZA
No sufficient improvement with finally fatal respiratory failure 2y after diagnosis
Abbreviations: δ, time between diagnosis of inflammatory bowel disease (IBD) and pulmonary manifestation (PM); y, years; mo, months; M, male; F, female; PFT, pulmonary function tests; (b), diagnosis of PM preceding IBD; XR, chest x-ray; CT-T, thoracic computed tomography; PRD, prednisolone; MP, methylprednisolone; IFX, infliximab; MES, mesalamine; 6-MP, 6-mercaptopurine.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Schmid, F.; Chao, C.-M.; Däbritz, J. Pathophysiological Concepts and Management of Pulmonary Manifestation of Pediatric Inflammatory Bowel Disease. Int. J. Mol. Sci. 2022, 23, 7287. https://doi.org/10.3390/ijms23137287

AMA Style

Schmid F, Chao C-M, Däbritz J. Pathophysiological Concepts and Management of Pulmonary Manifestation of Pediatric Inflammatory Bowel Disease. International Journal of Molecular Sciences. 2022; 23(13):7287. https://doi.org/10.3390/ijms23137287

Chicago/Turabian Style

Schmid, Florian, Cho-Ming Chao, and Jan Däbritz. 2022. "Pathophysiological Concepts and Management of Pulmonary Manifestation of Pediatric Inflammatory Bowel Disease" International Journal of Molecular Sciences 23, no. 13: 7287. https://doi.org/10.3390/ijms23137287

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop