eISSN: 1897-4317
ISSN: 1895-5770
Gastroenterology Review/Przegląd Gastroenterologiczny
Current issue Archive Manuscripts accepted About the journal Editorial board Abstracting and indexing Subscription Contact Instructions for authors Ethical standards and procedures
Editorial System
Submit your Manuscript
SCImago Journal & Country Rank
2/2023
vol. 18
 
Share:
Share:
Review paper

Biological markers of disease activity in inflammatory bowel diseases

Edyta Szymanska
1
,
Sylwia Szymanska
1
,
Maciej Dadalski
1
,
Jaroslaw Kierkus
1

1.
Department of Gastroenterology, Hepatology, Feeding Disorders, and Paediatrics, The Children’s Memorial Health Institute, Warsaw, Poland
Gastroenterology Rev 2023; 18 (2): 141–147
Online publish date: 2023/07/27
Article file
- Biological markers.pdf  [0.11 MB]
Get citation
 
PlumX metrics:
 

Introduction

Inflammatory bowel diseases (IBD) including Crohn’s disease (CD) and ulcerative colitis (UC) are chronic, relapsing intestinal conditions with globally increasing incidence, especially in developed countries [1, 2].

The pathogenesis of IBD, although still unknown, is most probably multifactorial and includes genetics, environmental factors, dysbiosis, and an impaired immune system [3, 4]. The diagnosis is based on both clinical and endoscopic features, and histopathology is the gold diagnostic standard [5]. However, endoscopy has notable disadvantages such as invasiveness, cost, and inconvenience, which limit its use for frequent monitoring of patients with IBD.

Fast, reliable, and non-invasive biological markers of intestinal inflammation and gut permeability have gained a lot of interest in recent years. They help to discriminate between functional and organic conditions, and as well as diagnosis they are used for monitoring disease activity when the diagnosis is already established, e.g. in IBD [6].

Stool biomarkers have higher specificity for intestinal inflammation compared to blood or serum markers. So far, faecal calprotectin (FCP) is the only biomarker approved and recommended by the European Crohn’s and Colitis Organization (ECCO).

Nonetheless, new biological markers such as faecal and serum zonulin, elafin, lipocalin, or haptoglobin have already been investigated in IBD [7, 8].

The aim of this review paper was to summarize new potential biological markers of inflammation for IBD and future approaches in their diagnosis and monitoring.

Serum and blood markers

Inflammation in IBD leads to an acute phase response detectable in the serum and blood, characterized by increased concentration of proteins involved in coagulation and fibrinolysis, such as fibrinogen and plasminogen; complement system components; proteinase inhibitors, including α1-antitrypsin, α1-anti-chymotrypsin; transport proteins such as haptoglobin and ceruloplasmin; and a variety of other proteins such as C-reactive protein (CRP) or ferritin [9]. Inflammatory cascade is mediated by serum proinflammatory cytokines, which are also elevated. These may include tumour necrosis factor-α (TNF-α), interferon-β, transforming growth factor-β, and interleukins (IL)-1β, IL-6, IL-8, IL-12, IL-17, and IL-23 [10].

Cellular components of blood may also indicate inflammation, which is reflected in elevations of white blood cell and platelet counts [10].

The erythrocyte sedimentation rate (ESR) is an indirect measure of inflammation, especially in chronic diseases. The test measures how quickly erythrocytes, or red blood cells, separate from a blood sample. Inflammation or cell damage make the red blood cells clump together, which makes them heavier, so they settle faster. The faster the red blood cells settle and fall, the higher the ESR [11].

Few blood or serum markers of inflammation have been validated in IBD, and CRP and ESR are the most widely available and used.

Serological markers

Various serologic tests have been proposed to differentiate between CD and UC; perinuclear antineutrophil cytoplasmic antibody (pANCA) and anti-Saccharomyces cerevisiae antibody (ASCA) are the most notable ones [12, 13]. It has been reported that pANCA is present in 50% to 65% of patients with UC and 10% to 25% of patients with CD, while ASCA is present in 55–65% of patients with CD and 5–20% of patients with UC [14, 15]. In a meta-analysis, combinations of ASCA and pANCA distinguished CD from UC with 40–50% sensitivity and > 90% specificity [16]. However, there are no data on the utility of serological markers to differentiate UC from CC and IBD undifferentiated (IBDU) in children [17]. Serological markers may also be useful to predict a disease behaviour [18]. It has been reported that ASCA is associated with a more aggressive disease course in paediatric CD [19]. Although there is emerging evidence that combining serological markers may increase the accuracy of differentiation between the 2 forms of IBD or their behaviour, the perfect assay or combination of antibodies has not been discovered. They have several limitations, so clinicians must be aware of the evidence on serological markers, interpret them with caution, and always correlate with the clinical presentation.

Calprotectin

Faecal calprotectin, a leukocyte protein of the S100 family belonging to the group of acute phase reactant proteins found in the granulocytes, neutrophil cytosol, in monocytes and activated macrophages, has been used to assess and monitor disease activity, mucosal healing (MH), and disease recurrence in patients with IBD [20]. It has been established that a cut-off point of 50 µg/g discriminates patients with IBD from controls with 79.4% specificity and 91.9% sensitivity and better correlation with clinical activity than C-reactive protein (CRP) [21].

Several factors such as age, diet, use of nonsteroidal anti-inflammatory drugs, exercise, and the presence of blood or mucus in stools may influence the level of FCP [2225]. Therefore, it is not specific for any disease; it may only help in discrimination between functional and organic conditions.

Tibble et al. demonstrated that increased concentration of FCP identified patients with IBD who were at risk of early relapse, which means it is also a good predictor of disease flare [26]. In the study by Costa et al. FCP level higher than 150 µg/g in patients in clinical remission was associated with a 2-fold increase in the relapse risk in CD and a 14-fold increase in UC, which may suggest stronger predictive value of clinical relapse in UC than in CD [27].

Ferreiro-Iglesias et al. reported a significant decrease in FCP level after 4 weeks of treatment in patients with CD who had complete response to therapy but not in partial or nonresponders [19]. In the study by Kolho et al. on children with IBD, a decrease in FCP level and clinical improvement were observed in paediatric patients with active disease treated with steroids, but it rarely dropped to normal values [28]. These outcomes indicate that FCP may serve as a good predictor of the response to therapy, as well.

Many studies have demonstrated good correlation of FCP with endoscopic disease activity in UC and in CD [29, 30]. Additionally, FCP has been shown to predict histopathological remission in children and adults with a cut-off point of 174 µg/g [31, 32].

Zonulin

Zonulin is a 47-kDa human protein that reversibly modulates the intercellular tight junctions crucial for maintaining physiological processes in the intestine [33]. Their impaired function leads to increased permeability in the epithelial layer of the small intestine [34].

Increased serum/plasma zonulin levels have been found in celiac disease, type 1 and type 2 diabetes, in obesity-associated insulin resistance, and in IBD [3537]. However, both the mechanism of intestinal inflammation development and the correlation between faecal and serum zonulin levels are not clear [38]. There are limited data on the zonulin use in IBD, and the majority of them include adult patients [6, 7, 39].

A Czech study examining 40 patients with IBD for faecal and serum zonulin showed that both of them were elevated in patients with active CD but not in UC. This finding may be explained by the fact that zonulin is considered as the best marker of increased permeability in the small intestine, and CD can extend to the whole gastrointestinal tract, including the small intestine [40]. Thus, zonulin levels may be higher in CD than in UC, which is restricted to the large intestine (with the exception of rare backwash colitis).

In a study by Caviglia et al. that investigated zonulin in patients with IBD and the correlation between its serum and faecal levels, serum concentrations were higher in IBD patients compared to the control group (34.5 (26.5–43.9) ng/ml vs. 8.6 (6.5–12.0) ng/ml, p < 0.001), but no correlation was observed between serum and faecal zonulin (rs = 0.15, p = 0.394) [8].

On the other hand, Wegh et al., who investigated which markers were most relevant to assess intestinal permeability in patients with UC, demonstrated that serum not faecal zonulin levels were elevated in active disease and had better correlation with other inflammatory markers such as CRP [39].

Our team analysed both serum and faecal zonulin in children with IBD. We demonstrated that faecal zonulin levels were elevated in those patients. Moreover, increased faecal zonulin was associated with CD activity and strongly correlated with FCP [41]. In the study investigating zonulin and I-FABP in our patients and their correlation with FCP, the only observed correlation was between faecal zonulin and FCP, and the strongest one was in CD: CD – R = 0.73, UC – R = 0.67, All – R = 0.67, CG – R = 0.65 [42].

Based on the outcomes from the above-mentioned studies, it seems that faecal zonulin may serve as another – along with FCP – biomarker of intestinal inflammation/increased intestinal permeability in IBD, especially in CD, both in adults and children.

Haptoglobin

Haptoglobin (HP) is a haemoglobin-binding protein with immunomodulatory properties.

It has been shown to have a protective effect against experimentally induced colitis – HP knockout mice presented with more weight loss and higher macroscopic and histological scores as compared with the wild-type ones [43]. This means that HP may play an important modulatory and protective role in inflammatory colitis in experimental models. Clinical data are contradictory. Maza et al. demonstrated that HP11 was significantly less common in CD, while Papp et al. showed its higher frequency in CD [44, 45]. However, in a well-powered study from the Marques group HP2 was found to be a risk allele for IBD, with a higher frequency in both CD and UC compared with controls [46]. Mouse model studies have demonstrated that Hp knockout mice are more susceptible to experimentally induced colitis than their wild-type littermates, which supports the protective effect of the HP1 allele in IBD. However, more studies are necessary to draw any firm conclusions on the role of HP in IBD.

Elafin

Elafin is one of the host defence peptides (HDPs) that has antimicrobial and antiprotease properties [47]. Initially, elafin was isolated from psoriatic skin, but it is also synthesized by epithelial cells of the gastrointestinal tract, lungs, or female reproductive system and inflammatory cells, including neutrophils, mast cells, and macrophages [48, 49].

Until now, there have been only a few studies investigating elafin in IBD, and their results are contradictory [5053].

Wang et al. demonstrated that the level of serum elafin was significantly elevated in patients with UC compared with the control group, while in patients with CD elafin was only mildly elevated, which was not statistically significant [52].

In the study by Schmid et al. elafin mRNA was expressed predominantly in the colonic biopsies of active UC [53].

On the other hand, Motta et al. reported that mucosal expression of elafin was decreased in patients with IBD [54]. Also, in the study by Zhang et al. expression of elafin mRNA in peripheral blood leukocytes was significantly decreased in patients with active UC but increased in UC remission. No significant differences in elafin mRNA were observed between patients with CD and controls [51]. Such outcomes – downregulation of elafin in IBD patients – may suggest that protease/antiprotease imbalance may take part in IBD development or may be a consequence of chronic inflammation leading to the destruction of epithelial cells, which are the main source of elafin.

A study from Poland investigating the role of elafin in the pathophysiology of inflammation in paediatric IBD has demonstrated that serum elafin was increased in the active phases of both UC and CD, but only in the remission of UC [55]. Based on the data published so far, elafin appears to be a potential candidate for a biomarker of UC.

Lactoferrin

Lactoferrin is an iron-binding glycoprotein secreted from glandular epithelial cells, and it can be identified in secretions overlying most mucosal surfaces including saliva, tears, vaginal secretions, faeces, synovial fluid, and mammalian breast milk [56]. Lactoferrin also has antibacterial activity [57]. Several reports have demonstrated that lactoferrin might be a primary factor in an acute inflammatory process because it is a major component of the secondary granules of neutrophils; its levels quickly increase during inflammation [58].

Sherwood reviewed the utility of lactoferrin in differentiating IBD from IBS, and showed that sensitivities for lactoferrin ranged from 78% to 88% and specificities from 85% to 100% [59]. However, there are contradictory data on the utility of lactoferrin in IBD.

Caccaro et al. demonstrated that lactoferrin distinguished between organic and functional disease with a sensitivity of 88% and a specificity of 94% [60]. Sugi et al. investigated lactoferrin, polymorphonuclear leukocytes (PMN)-elastase, lysozyme, and myeloperoxidase in stool and whole gut lavage fluid from patients with IBD and found that lactoferrin was a superior marker of intestinal inflammation [61]. In a further study, Langhorst et al. showed that lactoferrin and calprotectin differentiated active IBD from inactive IBD and IBS in 80% of cases, compared with 74% for PMN-elastase and 64% for CRP [62]. Judd et al. compared faecal concentrations of calprotectin, lactoferrin, and S100A12 in children with IBD, and they reported that calprotectin and lactoferrin correlated strongly with each other [63].

On the other hand, Silberer et al. reported that among a number of studied biomarkers, only PMN-elastase and calprotectin, but not lactoferrin, were useful to differentiate chronic IBD from IBS [64]. Moreover, other studies indicated that combining lactoferrin and calprotectin does not provide additional benefit [65, 66].

The potential utility of faecal lactoferrin in predicting risk of relapse in IBD has been investigated. Although the study included a small sample size, the results indicate that elevation of faecal lactoferrin may occur prior to clinical flares [67]. In addition, paediatric studies have demonstrated that faecal lactoferrin is a sensitive and specific marker of inflammation in children with IBD; faecal lactoferrin correlated with both clinical disease activity and serum inflammatory markers [59].

Overall, it seems that faecal lactoferrin is a good marker of inflammation. Although faecal lactoferrin may provide similar information to FCP, it has some limitations compared with calprotectin, which limits its utility in IBD.

Lipocalin

Lipocalin-2 (LCN2), a member of the lipocalin superfamily, also known as 24p3 or neutrophil gelatinase-associated lipocalin (NGAL), is a secreted glycoprotein produced by various cells including neutrophiles, myeloid, and intestinal epithelial cells [68, 69]. LCN2 is strongly induced by pro-inflammatory stimulation, such as IL-1β, IL-22, or Toll-like receptor (TLR) activation, and is secreted into the gut lumen in high concentrations [70, 71]. De Bruyn et al. demonstrated that serum LCN2 correlates with endoscopic activity in both CD and UC [72].

Zollner et al. compared FCP and faecal LCN2 (FLCN) in a cohort of more than 130 IBD patients and a group of healthy controls, and found comparable ability of both biomarkers in distinguishing between active and non-active disease and between IBD patients and non-IBD controls [73].

Table I summarizes the available biomarkers.

Table I

Biomarkers used and studied in IBD

TypeBiomarker
Serum/blood
  • Coagulation and fibrinolysis proteins: fibrinogen, plasminogen

  • Complement system components

  • Proteinase inhibitors: α1-antitrypsin, α1-anti-chymotrypsin

  • Transport proteins: haptoglobin, ceruloplasmin

  • C-reactive protein (CRP)

  • Erythrocyte sedimentation rate (ESR)

  • Ferritin

  • Proinflammatory cytokines: TNF-α, interferon-β, transforming growth factor-β, and interleukin (IL)-1β, IL-6, IL-8, IL-12, IL-17, IL-23

  • Morphology: white blood cell, platelet counts

Serological markers
  • Perinuclear antineutrophil cytoplasmic antibody (pANCA)

  • Anti-Saccharomyces cerevisiae antibody (ASCA)

Faecal markers
  • Calprotectin

  • Zonulin

Other
  • Elafin

  • Lipocalin

  • Haptoglobin

  • Lactoferrin

Future approaches

The research investigate more specific, sensitive, and responsive markers of inflammation that may be helpful in IBD, and to do that gene expression arrays, and metabolomic and proteomic platforms are used.

RNA as a biomarker in IBD

Studies of the IBD transcriptome have focused on the pathogenesis of IBD and on differentiation between CD and UC. A small study of the transcriptome of colonocytes from patients with UC was able to differentiate active UC from normal control tissue or UC in remission [74].

Other research used microRNAs (miRNA) – small, short (18–25 nucleotides), noncoding, single-stranded RNA species. The role of miRNA in IBD is not fully understood, but their ability to interact with messenger RNA and silence expression makes them key regulators of a variety of cellular processes. In one study, RNA was isolated from colonic biopsies of patients with UC and RNA was isolated and run on an miRNA gene expression array. It was shown that increased expression of miR-20b and miR-98 were associated with active UC, while increased expression of miR-125b-1* and let-7e* were associated with UC in remission [75]. Moreover, a study comparing circulating miRNA species in children with CD to healthy controls and children with celiac disease showed that 24 miRNA species were elevated in CD [76]. In addition to good sensitivity and specificity for a diagnosis of CD, these disease-associated miRNA species decreased after 6 months of treatment, which suggests that they may be useful as noninvasive biomarkers of inflammation [62].

Metabolomics

Metabolic profiling holds promise in differentiating IBD from other conditions and CD from UC, as well in potentially measuring inflammation. A variety of methods, such as NMR-spectroscopy, liquid chromatography-mass spectrometry, gas chromatography, or selective ion flow tube mass spectroscopy, have been applied to biospecimens including colonic biopsies, urine, and stool [77, 78]. In one study, volatile organic compounds have been measured in breath, and the results indicated a unique “breathprint” in children with IBD [79].

Proteomics

Protein profiles in serum, plasma, or tissue may also be distinct in IBD. Various techniques have been applied to separate and identify protein species, and they include among others, 2-dimensional gel electrophoresis, liquid chromatography, isobaric tags for relative and absolute quantification (iTRAQ), or tandem mass spectrometry (MS/MS) [80].

Pilot studies of proteomic profiling suggest that these approaches may be useful in identifying serum proteins that differentiate IBD from non-IBD patients, and potentially also response to therapy [81, 82].

Conclusions

Until now, FCP has been the only biomarker approved and recommended by the ECCO; however, many other non-invasive markers have been studied in IBD, and faecal zonulin, elafin, and probably lactoferrin appear to be the most promising ones. The future approaches to distinguish between IBD and other conditions, and to monitor disease activity and response to treatment include RNA, metabolomics, and proteomics.

Conflict of interest

The authors declare no conflict of interest.

References

1 

Flynn S, Eisenstein S. Inflammatory bowel disease presentation and diagnosis. Surg Clin North Am 2019; 99: 1051-62.

2 

Malik TA. Inflammatory bowel disease: historical perspective, epidemiology, and risk factors. Surg Clin North Am 2015; 95: 1105-22.

3 

Zhang YZ, Li YY. Inflammatory bowel disease: pathogenesis. World J Gastroenterol 2014; 20: 91-9.

4 

Geremia A, Biancheri P, Allan P, et al. Innate and adaptive immunity in inflammatory bowel disease. Autoimmun Rev 2014; 13: 3-10.

5 

Torres J, Bonovas S, Doherty G, et al. ECCO Guidelines on therapeutics in Crohn’s disease: medical treatment. J Crohns Colitis 2020; 14: 4-22.

6 

Piton G, Capellier G. Biomarkers of gut barrier failure in the ICU. Curr Opin Crit Care 2016; 22: 152-60.

7 

Ricciuto A, Griffiths AM. Clinical value of fecal calprotectin. Crit Rev Clin Lab Sci 2019; 56: 307-20.

8 

Caviglia GP, Dughera F, Ribaldone DG, et al. Serum zonulin in patients with inflammatory bowel disease: a pilot study. Minerva Med 2019; 110: 95-100.

9 

Vermeire S, Van Assche G, Rutgeerts P. Laboratory markers in IBD: useful, magic, or unnecessary toys? Gut 2006; 55: 426-31.

10 

Cioffi M, Rosa AD, Serao R, et al. Laboratory markers in ulcerative colitis: current insights and future advances. World J Gastrointest Pathophysiol 2015; 6: 13-22.

11 

Mendoza JL, Abreu MT. Biological markers in inflammatory bowel disease: practical consideration for clinicians. Gastroenterologie Clinique Et Biologique 2009; 33: S158-73.

12 

Mitchell PJ, Rabau MY, Haboubi NY. Indeterminate colitis. Tech Coloproctol 2007; 11: 91-6.

13 

Lewis JD. The utility of biomarkers in the diagnosis and therapy of inflammatory bowel disease. Gastroenterology 2011; 140: 1817-26.

14 

Kekilli M, Beyazit Y, Tas A, et al. Atypical pANCA as a marker of indeterminate colitis for the prediction of ulcerative colitis and Crohn’s disease. Wien Klin Wochenschr 2013; 125: 279-82.

15 

Plevy S. Do serological markers and cytokines determine the indeterminate? J Clin Gastroenterol 2004; 38: 51-6.

16 

Mokhtarifar A, Ganji A, Sadrneshin M, et al. Diagnostic value of ASCA and atypical p-ANCA in differential diagnosis of inflammatory bowel disease. Middle East J Dig Dis 2013; 5: 93-7.

17 

Laass MW, Ziesmann J, de Laffolie J, et al. anti-proteinase 3 antibodies as a biomarker for ulcerative colitis and primary sclerosing cholangitis in children. J Pediatr Gastroenterol Nutr 2022; 74: 463-70.

18 

Zhou G, Song Y, Yang W, et al. ASCA, ANCA, ALCA and many more: are they useful in the diagnosis of inflammatory bowel disease? Dig Dis 2016; 34: 90-7.

19 

Duarte-Silva M, Afonso PC, de Souza PR, Reappraisal of antibodies against Saccharomyces cerevisiae (ASCA) as persistent biomarkers in quiescent Crohn’s disease. Autoimmunity 2019; 52: 37-47.

20 

Pang T, Leach ST, Katz T, et al. Fecal biomarkers of intestinal health and disease in children. Front Pediatr 2014; 2: 6.

21 

Xiang JY, Ouyang Q, Li GD, Xiao NP. Clinical value of fecal calprotectin in determining disease activity of ulcerative colitis. World J Gastroenterol 2008; 14: 53-7.

22 

Poullis A, Foster R, Shetty A, et al. Bowel inflammation as measured by fecal calprotectin: a link between lifestyle factors and colorectal cancer risk. Cancer Epidemiol Biomarkers Prev 2004; 13: 279-84.

23 

Garcia-Planella E, Mañosa M, Chaparro M, et al, PRECUCAL study group Investigators. Serial semi-quantitative measurement of fecal calprotectin in patients with ulcerative colitis in remission. Scand J Gastroenterol 2018; 53: 152-7.

24 

Kolho KL, Alfthan H, Hämäläinen E. Effect of bowel cleansing for colonoscopy on fecal calprotectin levels in pediatric patients. J Pediatr Gastroenterol Nutr 2012; 55: 751-3.

25 

Calafat M, Cabré E, Mañosa M, et al. High within-day variability of fecal calprotectin levels in patients with active ulcerative colitis: what is the best timing for stool sampling? Inflamm Bowel Dis 2015; 21: 1072-6.

26 

Tibble JA, Sigthorsson G, Bridger S, et al. Surrogate markers of intestinal inflammation are predictive of relapse in patients with inflammatory bowel disease. Gastroenterology 2000; 119: 15-22.

27 

Costa F, Mumolo MG, Ceccarelli L, et al. Calprotectin is a stronger predictive marker of relapse in ulcerative colitis than in Crohn’s disease. Gut 2005; 54: 364-8.

28 

Kolho KL, Raivio T, Lindahl H, Savilahti E. Fecal calprotectin remains high during glucocorticoid therapy in children with inflammatory bowel disease. Scand J Gastroenterol 2006; 41: 720-5.

29 

D’Haens G, Ferrante M, Vermeire S, et al. Fecal calprotectin is a surrogate marker for endoscopic lesions in inflammatory bowel disease. Inflamm Bowel Dis 2012; 18: 2218-24.

30 

Szymańska E, Meglicka M, Dądalski M, et al. The level of faecal calprotectin as a noninvasive biomarker of mucosal healing in children with ulcerative colitis. Gastroenterology Rev 2020; 15: 343-8.

31 

Canani RB, Terrin G, Rapacciuolo L, et al. Faecal calprotectin as reliable non-invasive marker to assess the severity of mucosal inflammation in children with inflammatory bowel disease. Dig Liver Dis 2008; 40: 547-53.

32 

Guardiola J, Lobatón T, Rodríguez-Alonso L, et al. Fecal level of calprotectin identifies histologic inflammation in patients with ulcerative colitis in clinical and endoscopic remission. Clin Gastroenterol Hepatol 2014; 12: 1865-70.

33 

Fasano A. All disease begins in the (leaky) gut: role of zonulin-mediated gut permeability in the pathogenesis of some chronic inflammatory diseases. F1000Res 2020; 9: F1000 Faculty Rev-69.

34 

Fasano A. Intestinal permeability and its regulation by zonulin: diagnostic and therapeutic implications. Clin Gastroenterol Hepatol 2012; 10: 1096-100.

35 

Fasano A, Not T, Wang W. Zonulin, a newly discovered modulator of intestinal permeability, and its expression in coeliac disease. Lancet 2000; 29: 1518-9.

36 

Kort S, Keszthelyi D, Masclee AA. Leaky gut and diabetes mellitus: what is the link? Obes Rev 2011; 12: 449-58.

37 

Küme T, Acar S, Tuhan HA. The relationship between serum zonulin level and clinical and laboratory parameters of childhood obesity. J Clin Res Pediatr Endocrinol 2017; 9: 31-8.

38 

Fasano A. Intestinal zonulin: open sesame! Gut 2001; 49: 159-62.

39 

Wegh CAM, de Roos NM, Hovenier R, et al. Intestinal permeability measured by urinary sucrose excretion correlates with serum zonulin and faecal calprotectin concentrations in UC patients in remission. J Nutr Metab 2019; 2019: 2472754.

40 

Fasano A. Zonulin and its regulation of intestinal barrier function: the biological door to inflammation, autoimmunity, and cancer. Physiol Rev 2011; 91: 151-75.

41 

Szymanska E, Wierzbicka A, Dadalski M, Kierkus J. Fecal zonulin as a noninvasive biomarker of intestinal permeability in pediatric patients with inflammatory bowel diseases-correlation with disease activity and fecal calprotectin. J Clin Med 2021; 10: 3905.

42 

Szymanska E, Bierla J, Dadalski M, et al. New non-invasive biomarkers of intestinal inflammation and increased intestinal permeability in pediatric inflammatory bowel diseases and their correlation with fecal calprotectin: a pilot study. Minerva Gastroenterol 2022 Apr 19. doi: 10.23736/S2724-5985.22.03156-4.

43 

Márquez L, Shen C, Cleynen I, et al. Effects of haptoglobin polymorphisms and deficiency on susceptibility to inflammatory bowel disease and on severity of murine colitis. Gut 2012; 61: 528-34.

44 

Maza I, Miller-Lotan R, Levy AP, et al. The association of Haptoglobin polymorphism with Crohn’s disease in Israel. J Crohns Colitis 2008; 2: 214-8.

45 

Papp M, Lakatos PL, Palatka K, et al. Hungarian IBD Study Group Haptoglobin polymorphisms are associated with Crohn’s disease, disease behavior, and extraintestinal manifestations in Hungarian patients. Dig Dis Sci 2007; 52: 1279-84.

46 

Scheffler L, Crane A, Heyne H, et al. Widely used commercial ELISA does not detect precursor of haptoglobin2, but recognizes properdin as a potential second member of the zonulin family. Front Endocrinol 2018; 9: 22.

47 

Wehkamp J, Schmid M, Stange EF. Defensins and other antimicrobial peptides in inflammatory bowel disease. Curr Opin Gastroenterol 2007; 23: 370-8.

48 

Wiedow O, Schroder JM, Gregory H, et al. Elafin: an elastase-specific inhibitor of human skin. Purification, characterization, and complete amino acid sequence. J Biol Chem 1990; 265: 14791-5.

49 

Shaw L, Wiedow O. Therapeutic potential of human elafin. Biochem Soc Trans 2011; 39: 1450-4.

50 

Flach CF, Eriksson A, Jennische E, et al. Detection of elafin as a candidate biomarker for ulcerative colitis by whole-genome microarray screening. Inflamm Bowel Dis 2006; 12: 837-42.

51 

Zhang W, Teng G, Wu T, et al. Expression and clinical significance of elafin in inflammatory bowel disease. Inflamm Bowel Dis 2017; 23: 2134-41.

52 

Wang J, Ortiz C, Fontenot L, et al. High circulating elafin levels are associated with Crohn’s disease-associated intestinal strictures. PLoS One 2020; 15: e0231796.

53 

Schmid M, Fellermann K, Fritz P, et al. Attenuated induction of epithelial and leukocyte serine antiproteases elafin and secretory leukocyte protease inhibitor in Crohn’s disease. J Leukoc Biol 2007; 81: 907-15.

54 

Motta JP, Bermúdez-Humarán LG, Deraison C, et al. Food-grade bacteria expressing elafin protect against inflammation and restore colon homeostasis. Sci Transl Med 2012; 4: 158ra144.

55 

Krawiec P, Pac-Kożuchowska E. Clinical significance of serum elafin in children with inflammatory bowel disease. Biomedicines 2022; 10: 3267.

56 

Baynes RD, Bezwoda WR. Lactoferrin and the inflammatory response. Adv Exp Med Biol 1994; 357: 133-41.

57 

Levay PF, Viljoen M. Lactoferrin: a general review. Haematologica 1995; 80: 252-67.

58 

Baveye S, Elass E, Mazurier J, et al. Lactoferrin: a multifunctional glycoprotein involved in the modulation of the inflammatory process. Clin Chem Lab Med 1999; 37: 281-6.

59 

Sherwood RA. Faecal markers of gastrointestinal inflammation. J Clin Pathol 2012; 65: 981-5.

60 

Caccaro R, D’Inca R, Martinato M, et al. Fecal lactoferrin and intestinal permeability are effective non-invasive markers in the diagnostic work-up of chronic diarrhea. Biometals 2014; 27: 1069-76.

61 

Sugi K, Saitoh O, Hirata I, Katsu K. Fecal lactoferrin as a marker for disease activity in inflammatory bowel disease: comparison with other neutrophil-derived proteins. Am J Gastroenterol 1996; 91: 927-34.

62 

Langhorst J, Elsenbruch S, Koelzer J, et al. Noninvasive markers in the assessment of intestinal inflammation in inflammatory bowel diseases: performance of fecal lactoferrin, calprotectin, and PMN-elastase, CRP, and clinical indices. Am J Gastroenterol 2008; 103: 162-9.

63 

Judd TA, Day AS, Lemberg DA, et al. Update of fecal markers of inflammation in inflammatory bowel disease. J Gastroenterol Hepatol 2011; 26: 1493-9.

64 

Silberer H, Kuppers B, Mickisch O, et al. Fecal leukocyte proteins in inflammatory bowel disease and irritable bowel syndrome. Clin Lab 2005; 51: 117-26.

65 

Schoepfer AM, Trummler M, Seeholzer P, et al. Accuracy of four fecal assays in the diagnosis of colitis. Dis Col Rectum 2007; 50: 1697-706.

66 

Schoepfer AM, Trummler M, Seeholzer P, et al. Discriminating IBD from IBS: comparison of the test performance of fecal markers, blood leukocytes, CRP, and IBD antibodies. Inflamm Bowel Dis 2008; 14: 32-9.

67 

Walker TR, Land ML, Kartashov A, et al. Fecal lactoferrin is a sensitive and specific marker of disease activity in children and young adults with inflammatory bowel disease. J Pediatr Gastroenterol Nutr 2007; 44: 414-22.

68 

Skerra A. Lipocalins as a scaffold. Biochim Biophys Acta 2000; 1482: 337-50.

69 

Kjeldsen L, Johnsen AH, Sengeløv H, Borregaard N. Isolation and primary structure of NGAL, a novel protein associated with human neutrophil gelatinase. J Biol Chem 1993; 268: 10425-32.

70 

Behnsen J, Jellbauer S, Wong CP, et al. The cytokine IL-22 promotes pathogen colonization by suppressing related commensal bacteria. Immunity 2014; 40: 262-73.

71 

Nielsen BS, Borregaard N, Bundgaard JR, et al. Induction of NGAL synthesis in epithelial cells of human colorectal neoplasia and inflammatory bowel diseases. Gut 1996; 38: 414-20.

72 

de Bruyn M, Arijs I, De Hertogh G, et al. Serum neutrophil gelatinase B-associated lipocalin and matrix metalloproteinase-9 complex as a surrogate marker for mucosal healing in patients with Crohn’s disease. J Crohns Colitis 2015; 9: 1079-87.

73 

Zollner A, Schmiderer A, Reider SJ, et al. Faecal biomarkers in inflammatory bowel diseases: calprotectin versus lipocalin-2-a comparative study. J Crohns Colitis 2021; 15: 43-54.

74 

Bjerrum JT, Hansen M, Olsen J, et al. Genome-wide gene expression analysis of mucosal colonic biopsies and isolated colonocytes suggests a continuous inflammatory state in the lamina propria of patients with quiescent ulcerative colitis. Inflamm Bowel Dis 2010; 16: 999-1007.

75 

Coskun M, Bjerrum JT, Seidelin JB, et al. miR-20b, miR-98, miR-125b-1*, and let-7e*as new potential diagnostic biomarkers in ulcerative colitis. World J Gastroenterol 2013; 19: 4289-99.

76 

Zahm, AM, Thayu M, Hand NJ, et al. Circulating microRNA is a biomarker of pediatric Crohn disease. J Pediatr Gastroenterol Nutr 2011; 53: 26-33.

77 

Kohashi M, Nishiumi S, Ooi M, et al. A novel gas chromatography mass spectrometry-based serum diagnostic and assessment approach to ulcerative colitis. J Crohns Colitis 2014; 8: 1010-21.

78 

Storr M, Vogel HJ, Schicho R. Metabolomics: is it useful for inflammatory bowel diseases? Curr Opin Gastroenterol 2013; 29: 378-83.

79 

Patel N, Alkhouri N, Eng K, et al. Metabolomic analysis of breath volatile organic compounds reveals unique breath prints in children with inflammatory bowel disease: a pilot study. Aliment Pharmacol Ther 2014; 40: 498-507.

80 

Han NY, Kim EH, Choi J, et al. Quantitative proteomic approaches in biomarker discovery of inflammatory bowel disease. J Dig Dis 2012; 13: 497-503.

81 

Meuwis MA, Fillet M, Lutteri L, et al. Proteomics for prediction and characterization of response to infliximab in Crohn’s disease: a pilot study. Clin Biochem 2008; 41: 960-7.

82 

Li N, Wang XM, Zhang YF, et al. Comparative proteomics analysis of serum proteins in ulcerative colitis patients. Mol Biol Rep 2012; 39: 5659-67.

Copyright: © 2023 Termedia Sp. z o. o. This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 4.0 International (CC BY-NC-SA 4.0) License (http://creativecommons.org/licenses/by-nc-sa/4.0/), allowing third parties to copy and redistribute the material in any medium or format and to remix, transform, and build upon the material, provided the original work is properly cited and states its license.
 
Quick links
© 2024 Termedia Sp. z o.o.
Developed by Bentus.