SciELO - Scientific Electronic Library Online

 
vol.107 número2Etiología infecciosa de las diarreas estudiadas en un hospital de tercer nivel durante un periodo de cinco añosSarcoma de Ewing duodenal: localización inusual y traslocación atípica EWRS-1 índice de autoresíndice de materiabúsqueda de artículos
Home Pagelista alfabética de revistas  

Servicios Personalizados

Revista

Articulo

Indicadores

Links relacionados

  • En proceso de indezaciónCitado por Google
  • No hay articulos similaresSimilares en SciELO
  • En proceso de indezaciónSimilares en Google

Compartir


Revista Española de Enfermedades Digestivas

versión impresa ISSN 1130-0108

Rev. esp. enferm. dig. vol.107 no.2 Madrid feb. 2015

 

REVIEW

 

Advances in knowledge on microscopic colitis: From bench to bedside

Avance en el conocimiento de la colitis microscópica: desde el laboratorio a la cabecera de la cama

 

 

Danila Guagnozzi1 and Alfredo J. Lucendo2

1 Department of Gastroenterology. Hospital Dr José Molina Orosa. Lanzarote. Spain
2 Department of Gastroenterology. Hospital General de Tomelloso. Tomelloso, Ciudad Real. Spain

Correspondence

 

 


ABSTRACT

Microscopic colitis (MC) is a general term that describes a family of chronic inflammatory bowel diseases, including lymphocytic colitis (LC) and collagenous colitis (CC). The two forms are characterized by chronic watery diarrhea with normal or near normal endoscopic colonic appearance and specific histopathological abnormalities. Data from recent epidemiological studies reported the diagnosis of MC from several different regions in the world, providing that it can be a worldwide condition. The etiopathogenesis of MC still remains unknown but it is generally accepted that MC is a multifactorial disease, probably secondary to an abnormal immune reaction in predisposed individuals, triggered by different luminal factors (infections, drugs, autoimmunity and/or bile acids). Furthermore, some studies show that the epithelial barrier function in the colonic mucosa of MC patients is also impaired. Several mucosal factors of intestinal inflammation have been studied in MC, postulating that an aberrant T-lymphocyte response may lead to a chronic gut inflammatory condition, with the infiltration of colonic mucosa by different proportion of subset of T-lymphocytes. Little is known about the specific inflammatory mediators in MC pathogenesis, but a predominant Th1 type cytokine profile has been demonstrated. Currently, a number of medical treatments have been studied in MC patients, following mainly an empirical treatment approach. Further studies are needed in order to obtain prospective and more evidence-based data. In the future, it will be possible to develop causal treatment approaches after better understanding the molecular mechanisms behind the origin of the disease.

Key words: Microscopic colitis. Lymphocytic colitis. Collagenous colitis. Pathogenesis. Therapeutics. T-lymphocytes.


List of abbreviations:
MC: Microscopic colitis;
LC: Lymphocytic colitis;
CC: Collagenous colitis;
IBS: Irritable bowel syndrome;
BAM: Bile acid malabsorption;
iNOS: Inducible nitric oxide synthase;
NO: Nitric oxide;
EC: Enterochromaffin cells;
CTLs: Cytotoxic T lymphocytes;
IL-2: Interleukin-2;
Treg: Regulatory T-cells;
IL-10: Interleukin-10;
TGF-beta: Transforming growth factor-beta;
IFN-gama: Interferon-gama;
TNF-alpha: Tumor necrosis factor-alpha;
NF-KB: Nuclear factor-KB;
IL-17: Interleukin-17;
HLA: Human leucocyte antigen;
CD: Celiac disease;
IL-6: Interleukin-6;
IL-1b: Interleukin-1b;
IL-1RA: Interleukin-1RA;
5-ASA: 5-aminosalycilic acid.

 

Introduction

Microscopic colitis (MC) is a general term, used from 1980 (1) that describes a family of chronic inflammatory bowel diseases, including the two main entities, lymphocytic colitis (LC) and collagenous colitis (CC). The two forms are characterized by chronic or intermittent watery diarrhea, with normal o near normal endoscopic colonic appearance and specific microscopic abnormalities in the colonic mucosa, which allow to distinguish one form to the other. It is not clear whether LC and CC are the same disease entity, or have to be considered as different stages of the same condition (2,3). MC is a fairly new disease that is not always considered in patients with watery chronic diarrhea, because the diagnosis of MC may still be overlooked or misdiagnosed as irritable bowel syndrome (IBS). Epidemiological studies show that MC is diagnosed in up to 10 % of patients investigated for chronic or intermittent watery non-bloody diarrhea and in more than 20 % of patients older than 70 year old, being more frequent in elderly women and smokers (4). Data from recent epidemiological studies reported the diagnosis of MC from several different regions in the world, making it a worldwide condition. Nowadays, after a period of generally rising incidence of MC, data from recent epidemiological studies, show incidence rates ranging between < 1/100.000 to 10.8/100.000 inhabitants for CC and between 2.1/100.000 to 18.9/100.000 inhabitants for LC (5-19) (Table I). The pathogenesis of MC still remains unknown, but it is considered to be multifactorial, showing the importance of an impaired mucosal barrier function and the interaction between luminal content (including from bacteria to drugs) and the mucosal intestinal immune system. A better understanding of the immunological basis of inflammation in the colonic mucosa could lead to better therapeutic options and the design of new strategies to treat MC patients in the future.

 

 

Methods

This paper reviews the current evidences on the pathogenetic mechanisms underlying MC and its implications in the therapeutic management of the disease. We searched the PubMed, Cochrane, MEDLINE, and Scopus libraries, using the following individual and combined key words: Microscopic colitis; collagenous colitis; lymphocytic colitis; treatment or therapy; etiology; pathophysiology; genetic factors; luminal factors; epithelial barrier function; mediators of intestinal inflammation; budesonide; mesalazine; loperamide; bismuth subsalicylate; probiotics; immunosuppressive therapy; anti-TNF treatment. Reference lists in the articles obtained were also searched in order to identify other potential sources of information. The results were limited only to studies published, written in English.

Etiopathogenetic mechanisms of MC

It is generally accepted that MC is a multifactorial disease, probably secondary to an abnormal immune reaction that appears in predisposed individuals, triggered by different luminal factors (infections, drugs, autoimmunity and/or bile acids). The mechanism by which the alteration of the mucosal immune response generates the dominant symptom of the disease (diarrhea) is still under investigation, with several data showing that the diarrhea in MC patients could have an inflammatory origin. Indeed, the severity of diarrhea seems to be associated with the intensity of inflammation in the lamina propria in LC, while in patients with CC is not correlated to the thickness of the collagenous band (20). Furthermore, in those patients with CC, who had a temporary ileostomy, the recurrence of inflammation in the lamina propria was the first histological change observed in the development of the symptomatic disease (21). Nonetheless, other data indicate that osmotic and secretory components could also contribute to the development of diarrhea in MC patients. In fact, fasting can reduce the diarrhea (22) and some authors have demonstrated that diarrhea can be originated from a reduced Na+ and Cl- absorption, together with an active chloride secretion (23,24). Recently, a study from colonic biopsies from patients with CC and LC has shown that electrogenic sodium transport diminished and the epithelial resistance decreased. Finally, unlike patients with IBS, there are no signs of visceral hypersensitivity in patients with CC, also showing a normal ano-rectal function, despite the presence of rectal inflammation (25). Therefore, a combination of several factors may contribute to the induction and maintenance of an inflammatory response in the colonic mucosa of patients with MC to produce the symptoms of the disease (Fig. 1), which we will describe in detail in the following sections.

 

 

Luminal factors

Several luminal factors have been studied as putative triggers, to induce an impaired inflammatory mucosa response in MC patients:

Firstly, an infectious etiology is proposed, despite no specific pathogen has been identified until now. Some studies show the onset of MC following a gastrointestinal infections, as in the case of a Clostridium difficile infection, that can catalyze the inflammatory response to induce the development of CC (26), or the observation that serum antibodies against to Yersinia enterocolitica were more common among CC patients compared to controls, leading to the speculation of a previous Yersinia enterocolitica infection triggering the development of the disease (27). A role for gastrointestinal infections in the pathogenesis of MC is strikingly resembling to the hypothesis of post-infectious IBS (28), with similar clinical presentation, spontaneous regression and similar histological changes (intraepithelial and submucosal infiltration by lymphocytes and mast cells) (29).

Secondly, it is widely known that colonic infusion of bile acids may determine colitis in animal models (30), and in patients with ileal resection, with subsequent bile acid malabsorption (BAM), may have diarrhea (31). Some small retrospective case series have found BAM to be presented in up to 60 % of patients with LC and in up to 44 % of those with CC (32,33). This suggests that, at least in some patients, BAM might have a key role in the development of the disease. However, the evidence on the role of bile acids in the pathogenesis of MC is still conflicting. In fact, small studies conducted with bile acid breath test showed little or no evidence of BAM in MC patients (34-36) and an association between cholecystectomy and MC has not been found (37). On the other hand, some studies investigating the retention of selenium homocholyltaurine have found variable degrees of BAM (31,38) and bile acid binding treatment, such as using cholestyramine, has been shown to be effective in CC patients. Finally, a recent study has shown that a low concentration of dihydroxy bile acids can aggravate the mucosal barrier dysfunction in CC patients by increasing the uptake of non-pathogenic bacteria (39). However, whether BAM is causative or not still remains questionable (40) and on the basis of the new evidence, BAM and MC are probably associated but independent diseases.

Thirdly, drug consumption has been considered as an environmental risk factor causing or triggering MC. The likelihood of a specific drug to trigger MC has been shown in an interesting paper (41); drug imputation was assessed by using a French algorithm and adding the bibliographic score, calculated by the number of previous reports of a similar drug-event association (42). Significant associations were reported between MC and certain drugs with a high likelihood in triggering the disease, including non-steroidal anti-inflammatory drugs (NSAIDs), proton pump inhibitors (PPIs), Ranitidine, Sertraline, Clozapine, Acarbose, Aspirin (ASA), Ticlopidine, Flavonoid vein tonic drugs and Entacapone (41). However, it is important to highlight that only by a re-challenge protocol (this is, assessing disease remission after drug withdrawal and recurrence after new expositions) it is possible to truly confirm causality relationships between some drugs and the development of a disease. Data from drug re-challenges are available only for few drugs, including Acarbose, Cyclo 3 forte, Ranitidine, Lansoprazole and Omeprazole (43-47), and some of them are limited to clinical recurrence, with no available histological assessment. Therefore, the possible causative nature of these associations is a matter of ongoing discussion, also because many drugs and their metabolites can produce diarrhea by several different mechanisms. They can affect the colon directly, either through their pharmacological activity, or through idiosyncratic hypersensitivity reactions, or indirectly by altering the colonization of gastrointestinal microorganisms (4). Several drugs thought to be associated with MC are also known to be involved in the development of chronic or recurrent diarrhea as a side effect (48), further confusing the possible causal role of drugs in the development of the disease. In this way the association between the development of MC and drug intake, could be considered as an idiosyncratic drug reaction and should be assessed before starting a specific treatment for MC.

Epithelial barrier dysfunction

The intestinal epithelium is a selective and regulated barrier that permits ions, water and nutrients to be absorbed, but restricts the passage of harmful molecules, bacteria, viruses and other pathogens (49). According with some studies, the colonic epithelial barrier function is impaired in MC patients, while the small bowel mucosa integrity remains still intact (50). In particular, we know that paracellular permeability is increased in CC patients, due to a reduced expression of occludin and claudin-4, which are important tight junction proteins, resulting in a reduction of the epithelial resistance (24). An in vitro study carried out over colonic biopsies from CC patients who were in clinical remission, found a significant dysfunction in the mucosal barrier associated with an increased transmucosal uptake of non-pathogenic bacteria. This worsened in case of an active disease and persisted despite effective treatment with budesonide (51). The diversion of the fecal stream by an ileostomy normalized the mucosal permeability and epithelial degeneration, in parallel with decreasing of the mucosal inflammation in CC patients. The altered mucosal permeability recurred after restoring the bowel continuity, and the thickened collagenous layer latter reappeared (52). Finally, increased levels of both inducible nitric oxide synthase (iNOS) and nitric oxide (NO) are also observed in the colonic mucosa of MC patients, which leads to an increased paracellular transit, by increasing the tight junction permeability (53-55). Some authors postulated that the increased levels of NO are caused by a higher secretion of serotonin from enterochromaffin (EC) cells, as observed in colonic mucosa samples from LC patients (56,57). The increase in colonic serotonin cell density probably results from the interaction between lymphocytes and EC cells. Serotonin accelerates the intestinal motility and promotes water and electrolyte secretion, with a secondary compensatory increased expression in peptide YY, as observed in LC patients (57). It in turn, inhibits prostaglandin E2 and vasoactive intestinal polypeptide activation, stimulating the absorption of water and electrolytes (58-60). This probably explains the intermittent evolution of the diarrhea in MC patients. However, it is not yet clear if mucosal barrier dysfunction is a primary or secondary phenomenon, underlying the mucosal inflammation (Fig. 2).

 

 

Impaired mucosal immune system

Several mucosal factors leading to intestinal inflammation have been studied in MC, postulating that an impaired adaptive immune response through aberrant T-cell responses leads to chronic gut inflammatory conditions (61). The number of CD3+ T-lymphocytes in the lamina propria and intraepithelial compartment is increased in LC and CC (62), as it is defined as a hallmark for these diseases. On the basis of immunohistochemical staining and flow cytometric analyses, most of the T lymphocytes are CD4+ within the lamina propria and CD8+ in intraepithelial compartment, carrying the alpha/beta form of the T-cell receptor (62), with a mixed Th17/Tc17 and Th1/Tc1 mucosal cytokine profile (63). The heavy infiltration by CD8+ cytotoxic T-lymphocytes (CTLs) in the colonic mucosa of MC patients has been recently shown to be due to an increased expansion of resident T-cells. It suggests that the activating antigen localizes in the colonic mucosa, such as demonstrated by a reduced T-cell receptor excision circles level with enhanced expression of Ki67+ T-cells in the colonic mucosa of MC patients (61,64). In fact, other studies have also demonstrated an elevated proportion of memory CD8+ T-cells (Ki67+ and CD45RO+ cells) in intraepithelial and lamina propria compartments, being more pronounced in CC than LC and in active disease, compared with the disease in histopathological remission state. It is important to highlight that compared to spleen memory T-cells, memory CD8+ T-cells can be more rapidly reactivated to produce uncontrolled or excessive cytotoxic activity against luminal antigens, developing and maintaining an intestinal chronic inflammatory response (64). Furthermore, CTLs induce proliferation of regulatory T-cells (Treg) through the secretion of interleukin (IL-2), which in turn inhibits CTLs-induced cytotoxic tissue damage, in order to maintain the self-tolerance status by the production of suppressor cytokines (IL-10, transforming growth factor [TGF]-beta) and by cell-to-cell contact-dependent mechanisms (65-68). CD25+FOXP3+ Treg are increased in the lamina propria of MC patients, probably blocking the spoiling effect of CTLs (69,70). In fact, this action is modulated by TGF-beta acting as a suppressive factor for inflammatory cells proliferation and function, with an additional role in causing the accumulation of collagen as observed in CC (71). Several additional factors are being studied as responsible causes in the formation of the thick subepithelial collagen band. Among others, increased numbers of activated subepithelial myofibroblasts (72,73) and vascular endothelial growth factor, which promote the accumulation of immature subepithelial matrix (74). In contrast, patients with LC have significant more CTLs than those with CC and less CD25+FOXP3 Treg than CC patients, although with increased levels compared to normal colonic mucosa (69). These observations support the theory that LC and CC could be considered as different stages of the same condition, with increased numbers of CTLs at the earliest stage (LC) and subepitelial collagen bands and increased CD25+FOXP3+ Treg in the later stage (CC) (69). Besides the lymphocytic infiltration, an increased number of eosinophils can be observed in the colonic mucosa of CC patients (75-77), along with increased levels of eosinophilic cationic protein (77) and degranulation of major basic protein (76), suggesting the involvement of the innate immune system in the pathogenesis of MC.

One report showed a predominant Th1-type cytokine profile, with marked increase in interferon (IFN)-gamma, IL-15 and iNOS expression and with lesser increase in tumor necrosis factor (TNF)-alpha (78). Increased activities of nuclear factor-KB (NF-KB), iNOS, IFN-gamma and COX-2, have been shown in other studies (79-82), especially in CC, where their increases were significantly higher than in LC. An increased expression of interleukin-17 (IL-17) in epithelial and inflammatory cells of MC patients has been recently demonstrated (63,83), which plays an important role in antimicrobial immunity at mucosal barriers and in the amplification of intestinal inflammatory responses.

Genetic factors

Whether genetic predisposition is of importance in MC is unclear. Several aspects in the sparse scientific evidences indicate that a genetic background could play a role in the etiology of MC. Familiar aggregation of MC cases were demonstrated in a Swedish study, which found five families with the occurrence of MC, with a sister-sister relationship, independently of smoking habits (84). Mother-daughter and father-son relationships have also been described (85,86).

Recently, a familial case of CC below 14 years old has been reported in a mother-daughter relationship (87). Unfortunately, until now, no concordance among twins has been reported for MC, pointing out that not only genetic predisposition, but also environmental factors, should also be considered in order to explain the familial occurrence of MC.

In addition, small studies have found an association between MC with various autoimmune-related human leukocyte antigens (HLA) haplotypes. An increasing prevalence of HLA-A1 and a decreased of HLA-A3 haplotypes has been found in LC, with no associated in CC patients (88). Some studies have reported on the association of MC with the HLA-DQ haplotypes predisposing to celiac disease (CD), with a prevalence level of HLA-DQ2 alleles up to 64 % of MC patients (among whom only 4 % had a concomitant CD), compared with only 31 % in control subjects (89). The association of HLA-DQ2 with LC without the association to CD has also been found (90).

Finally, different genetic polymorphisms have been studied in MC patients: the frequency of carriers for the GG allele of the SNP of matrix metalloproteinase was found significantly higher in CC patients than in healthy donors, being the odds ratio for an increased risk of CC 1,9 (95 % confidence interval [CI], 1, 3.5) (91). Unfortunately, the carrier frequencies of the alleles in SNP 8, 12 and 13 of NOD2/CARD15 gene were not different between CC patients and healthy blood donors (92). The IL-6-174 gene polymorphism has also been related with MC, showing that the IL-6 GG and G genotypes were more frequent in patients with MC compared to controls, being the odds ratio of 1.5 (95 % CI 1.041, 2.203) and 1.9 (95 % CI 1.078, 3.495), respectively. An association with other polymorphisms of cytokines IL-1b, IL-1RA, IL-10 and CD14 was not found in the same study (93). The genotypes GG and G of the IL-6 gene are associated with an increased IL-6 production that could contribute to the development of pro-inflammatory responses in the intestinal mucosa. Finally, an association of MC with HLA DR3-DQ2 haplotype and TNF2 allele has also been found (94). Further studies are needed in order to strength the knowledge on the role of genetic factors in the origin of MC.

 

From bench to bedside: treatment options for MC

The treatment of MC should take into account the severity of symptoms and their impact on the patients' quality of life, the efficacy of the drugs in obtaining clinical remission (accompanied or not with histological remission), and the secondary effects associated to short and long term treatments. Considering the high rate of spontaneous remission and sustained remission after short term therapy, the long-term therapy should be considered preferentially on relapsing or refractory patients, for whom an intermittent therapy should be favored (4). In recent years, a number of randomized controlled trials (RCT) have been provided for a more evidence-based approach to treat MC patients. More evidence is available on the treatment of CC than LC, but there is no reason to treat these two entities differently, because the drugs that seem to be effective in one tend to be effective also in the other disease. Several medical treatments have been studied in MC patients, following empirical approaches. In fact, currently there is no casual treatment approach for MC, although increasing data on the effects of some drugs over pathogenetic mechanisms involved in the development of MC are now available.

Induction of remission and maintenance treatment in patients with active MC

Budesonide

Budesonide is a non-halogenated gluco-corticosteroid with a very high receptor binding affinity and a limited systemic availability, without the significant adverse events that frequently occur in systemic corticosteroids-based treatment. Budesonide is currently the most promising treatment for MC; the strongest evidence on its efficacy, coming from several RCTs. It should be considered the first-line treatment in active MC (4). Three RCT in CC (95-97) and two in LC (98,99) have demonstrated that budesonide therapy (9 mg/day for 6-8 weeks) is effective in inducing the clinical remission and to improve the patients' health-related quality of life. Furthermore, in a recent study, budesonide was significantly superior to mesalazine for inducing clinical remission (80 % vs. 44 %, p = 0.0035) in CC patients (100). Two Cochrane Collaboration systematic reviews with meta-analysis reported a high response rate for achieving a clinical remission compared to placebo, with a pooled response rate of 81 % in CC (101) and an odds ratio of 9 in LC (102), with these last results also confirmed by other recent meta-analysis (103). However, a high rate (61-80 %) of relapse within 2 weeks after stopping the treatment was reported in all trials, which required studying the benefit of a maintenance treatment. In fact, two RCTs have shown that clinical remission and histological response can be maintained with budesonide at a dose of 6 mg/day for 6 months (104,105), and a mean dose of 4.5 mg/day maintained clinical remission for at least 1 year in 61.4 % in CC patients vs. only 16.7 % in the placebo group (p < 0.001), with few adverse drug reactions. As a whole, these results suggest the usefulness of maintenance treatment with a low-dose of budesonide, to prevent this high relapse rate (106).

Regarding the mechanism of action of this drug, we know that the glucocorticoid receptor is ubiquitously found in nearly all tissues and its interaction with the cells and mediators plays a key role in the inflammatory cascade, preventing epithelial dysfunction in vitro models of inflammation, by inhibiting the activation of both T-cells and monocytes (107), modulating the inflammatory regulator NF-KB (108) and acting by other different mechanisms (109,110). However, we still lack enough data about what are the exact mechanisms by which budesonide acts in order to produce its therapeutic effect in MC patients. From the few data available, we know that budesonide can modify the different lymphocyte subpopulations infiltrating the intestinal mucosa in MC patients, as showed by immunohistochemical staining and flow cytometric analysis (96,98,111). In fact, budesonide treatment is often associated with the restoration of histology in a majority of LC cases, and with a partial or complete normalization of the inflammatory infiltrate in 30.8 % and 69.2 % CC patients, respectively (112). In contrast, a recent study failed to demonstrate significant reductions in the proportion of total CD8+ intraepithelial T-cells, when T-cells from lamina propria were separately analyzed by flow cytometry in budesonide-treated LC and CC patients (64). In the same study, no differences in the proportion of CD45RO+ or Ki67+ cells, nor CD8+ or CD4+ intrahepitelial and lamina propria T-cells, between untreated and budesonide treated patients with MC were found, except a single LC treated patient showing a declined proportion of these cells. A declined proportion of CD4+8+ lamina propria and IELs cells were only found in CC treated patients, but not in LC patients (64). A reduction in the thickness of the subepithelial collagen band can be seen in treated patients with CC, but it seems to take a longer time than for the resolution of the inflammatory infiltrate (112). Moreover, budesonide treatment can reduce the iNOS mRNA expression that correlates with the inflammatory and the clinical activity of CC patients (113). Finally, the efficacy of budesonide in MC might be in part also due to an increase in the ileal reabsorption of bile acids (114). However, there are some pathogenetic mechanisms on which budesonide has no effect, as demonstrated by the persistent mucosal barrier dysfunction after budesonide treatment (51) despite of the fact of restoring the electrogenic sodium transport impairment (115) and the altered mucosal reactivity (51).

Mesalazine

5-Aminosalicylic acid (5-ASA) such as mesalazine has a local anti-inflammatory effect, as demonstrated also in other inflammatory bowel diseases. There are few evidence-based data about its usefulness in MC patients. In retrospective studies 5-ASA have been shown to be effective in about half of the patients at a dose of 2.4-3 g/day, with an effective rate ranging from 86 %-42 % in Spanish studies, to 35-42 % in American and other European studies (116-119). Prospective open-label studies showed one high efficacy of mesalazine for inducing remission in MC (73 % in CC and 85 % in LC), especially when combined with cholestyramine in CC patients (102,120). However, in the unique RCT published, mesalazine resulted less effective than budesonide and placebo to induce clinical remission in CC patients (100). Therefore, the current value of mesalazine for the treatment of MC patients remains unclear and further studies are needed.

Cholestyramine

Even when cholestiramine has never been assayed in a RCT, evidence from uncontrolled retrospective data shows that this bile acid binder (at a dose of 4 g/day) could be effective in MC, especially in CC patients with a concomitant BAM (121). As stated above, the benefit of cholestyramine is slightly better if combined with mesalazine, as showed in a prospective no placebo-controlled study (120); these data need to be confirmed to clarify the real therapeutic value of this drug. In fact, patients on bile acid binders had no significant changes in histopathology, despite a good effect on their symptoms (122). However, it is important to highlight that its beneficial effect is not only bile acid-specific, as cholestyramine also binds bacterial toxins, which might also contribute to reduce the symptoms in MC patients by a bile acids independent mechanism, which highlights the important role for luminal antigens in the pathogenesis of MC.

Other treatment options

Loperamide

Some drugs could be used to produce symptomatic effect such as loperamide, which does not achieve sustained clinical remission and produces no impact on colonic inflammation. However, loperamide could be reserved to control symptoms in mild cases of MC (123).

Probiotics

Probiotics are other treatment whose potential role in the induction and/or maintenance of clinical remission in MC should also be further investigated. Until now, they have been exclusively investigated in CC patients: In particular, Lactobacillus acidophilus LA-5, Bifidobacterium animalis subspp lactis BB12 and Boswellia serrata extracts did not show benefit, in terms of clinical and histological response or in improvement of quality of life, compared to placebo (124,125). Only the probiotic Escherichia coli strain Nissle 1917 has achieved in an open-label uncontrolled trial a reduction in stool frequency in 64 % of CC patients (126).

Bismuth derivatives

Bismuth subsalicylate, a derivative of salicylic acid with potential anti-inflammatory and antibacterial actions has occasionally been used in America on mild to moderate active MC: A RCT and an additional open-label study showed clinical and histological response in MC patients (127,128), but toxicity concerns have determined that this drug is not available and not used in many countries.

Therapeutical options for refractory MC patients and future prospects

Only a small subset of patients with MC are refractory to steroids treatment, amounting approximately less than 5 % in a population-based cohort study (129). In patients who failed to respond and in patients intolerant to budesonide, the use of immunosuppressive therapies should be considered, once confirmed the histological diagnosis of MC, carried out a correct differential diagnosis, ascertained the compliance to the treatment prescribed and the absence of exposures to high risk medications for MC. However, the experience with immunomodulators in MC is mainly anecdotal, with no RCT available until now, and with its use mainly derived because of the pathogenetic similarities between MC and inflammatory bowel diseases. In particular, Azathioprine or 6-Mercaptopurine, have been demonstrated useful in some MC patients refractory to budesonide with a response rate of 89 %, associating a steroid sparing effect. However, approximately 50 % of MC patients were intolerant to these drugs (129,131). Doses used in MC have been very similar to that of inflammatory bowel disease. Methotrexate (MTX) has also been studied in a retrospective report on CC patients, showing a beneficial effect with oral low-dose (5-25 mg). However, most patients in this report had not been previously treated with budesonide (132). In contrast, a prospective study conducted on patients intolerant or refractory to budesonide who received subcutaneous MTX (15-25 mg) showed no clinical remission (133).

An effective use of anti-TNF therapy (infliximab and adalimumab) has been reported in refractory severe MC patients, as a third-line treatment or "rescue therapy" to avoid surgery (diverting ileostomy or colectomy) in selected intractable cases (134,135). On a pathogenetic point of view, the TNF-alpha released by activated macrophages and T-cells triggers a chain of cellular mediators that causes secondary intestinal damage. Limited data have shown an increased expression of TNF-alpha in the colonic mucosa of MC patients (136,137), although at a very weak level (< 10 % in epithelial and inflammatory cells of MC patients) (83). A putative role for IL-17 in the pathogenesis of MC has been recently proposed, after documenting an increased expression in epithelial and inflammatory cells of MC patients (83). In fact, the regulation of IL-17 appears as a viable future therapeutic option in the treatment of MC patients, since a novel inhibitor of the production of IL-17 in activated lymphocytes (Vidofludimus) has been studied in animal models of chronic intestinal inflammation (138). The better understanding of the molecular mechanisms behind the development and maintenance of colonic inflammation in MC patients will make possible the development of future specific drugs to control disease activity. Evidence-based data deserve further prospective studies to obtain more conclusive data regarding this small subset of patients.

 

Conclusions

Knowledge about MC has constantly increased since the first description of the disease, to the point that CC and LC have been established as well-defined diseases. MC is currently considered as a worldwide condition, as common as inflammatory bowel disease. However its pathogenesis still remain unknown and the approach to the treatment of MC patients is mainly empirical and not causal, based on very comprehensive evidence. Fundamental advances in the understanding of the pathogenetic mechanisms of the development and maintenance of chronic intestinal inflammation in MC patients are needed, to further allow the development of novel and effective specific therapies.

 

References

1. Read NW, Krejs GJ, Read MG, Santa Ana CA, Morawski SG, Fordtran JS. Chronic diarrhea of unknown origin. Gastroenterology 1980;78:264-71.         [ Links ]

2. Perri F, Annese V, Pastore M, Andriulli A. Microscopic colitis progressed to collagenous colitis: A morphometric study. Ital J Gastroenterol 1996;28:147-51.         [ Links ]

3. Shaz BH, Reddy SI, Ayata G, Brien T, Farraye FA, Antonioli DA, et al. Sequential clinical and histopathological changes in collagenous and lymphocytic colitis over time. Mod Pathol 2004;17:395-401.         [ Links ]

4. Münch A, Aust D, Bohr J, Bonderup O, Fernández Bañares F, Hjortswang H, et al. Microscopic colitis: Current status, present and future challenges. J Crohns Colitis 2012;6:932-45.         [ Links ]

5. Bohr J, Tysk C, Eriksson S, Järnerot G. Collagenous colitis in Orebro, Sweden, an epidemiological study 1984-1993. Gut 1995;37:394-7.         [ Links ]

6. Olesen M, Eriksson S, Bohr J, Järnerot G, Tysk C. Microscopic colitis: A common diarrhoeal disease. An epidemiological study in Orebro, Sweden, 1993-1998. Gut 2004;53:346-50.         [ Links ]

7. Wickbom A, Bohr J, Eriksson S, Udumyan R, Nyhlin N, Tysk C. Stable incidence of collagenous colitis and lymphocytic colitis in Örebro, Sweden, 1999-2008: A continuous epidemiologic study. Inflamm Bowel Dis 2013;19:2387-93.         [ Links ]

8. Vigren L, Olesen M, Benoni C, Sjöberg K. An epidemiological study of collagenous colitis in southern Sweden from 2001-2010. World J Gastroenterol 2012;18:2821-6.         [ Links ]

9. Thörn M, Sjöberg D, Ekbom A, Holmström T, Larsson M, Nielsen AL, et al. Microscopic colitis in Uppsala health region, a population-based prospective study 2005-2009. Scand J Gastroenterol 2013;48:825-30.         [ Links ]

10. Agnarsdottir M, Gunnlaugsson O, Orvar KB, Cariglia N, Birgisson S, Bjornsson S, et al. Collagenous and lymphocytic colitis in Iceland. Dig Dis Sci 2002;47:1122-8.         [ Links ]

11. Fernández-Bañares F, Salas A, Forné M, Esteve M, Espinós J, Viver JM. Incidence of collagenous and lymphocytic colitis: A 5-year population-based study. Am J Gastroenterol 1999;94:418-23.         [ Links ]

12. Fernández-Bañares F, Salas A, Esteve M, Pardo L, Casalots J, Forné M, et al. Evolution of the incidence of collagenous colitis and lymphocytic colitis in Terrassa, Spain: A population-based study. Inflamm Bowel Dis 2011;17:1015-20.         [ Links ]

13. Guagnozzi D, Lucendo AJ, Angueira-Lapeña T, González-Castillo S, Tenias Burillo JM. Prevalence and incidence of microscopic colitis in patients with diarrhoea of unknown aetiology in a region in central Spain. Dig Liver Dis 2012;44:384-8.         [ Links ]

14. Bjornbak C, Engel PJH, Nielsen PL, Munck LK. Microscopic colitis: Clinical findings, topography and persistence of histopathological subgroups. Aliment Pharmacol Ther 2011;34:1225-34.         [ Links ]

15. Pardi DS, Loftus EV Jr, Smyrk TC, Kammer PP, Tremaine WJ, Schleck CD, et al. The epidemiology of microscopic colitis: A population based study in Olmsted County, Minnesota. Gut 2007;56:504-8.         [ Links ]

16. Gentile NM, Khanna S, Loftus EV Jr, Smyrk TC, Tremaine WJ, Harmsen WS, et al. The epidemiology of microscopic colitis in Olmsted County from 2002 to 2010: A population-based study. Clin Gastroenterol Hepatol 2014;12:838-42.         [ Links ]

17. Williams JJ, Kaplan GG, Makhija S, Urbanski SJ, Dupre M, Panaccione R, et al. Microscopic colitis-defining incidence rates and risk factors: A population-based study. Clin Gastroenterol Hepatol 2008;6:35-40.         [ Links ]

18. Andrews CN, Beck PL, Wilsack L, Urbanski SJ, Storr M. Evaluation of endoscopist and pathologist factors affecting the incidence of microscopic colitis. Can J Gastroenterol 2012;26:515-20.         [ Links ]

19. Stewart M, Andrews CN, Urbanski S, Beck PL, Storr M. The association of coeliac disease and microscopic colitis: A large population-based study. Aliment Pharmacol Ther 2011;33:1340-9.         [ Links ]

20. Olesen M, Eriksson S, Bohr J, Järnerot G, Tysk C. Microscopic colitis: A common diarrhoeal disease. An epidemiological study in Orebro, Sweden, 1993-1998. Gut 2004;53:346-50.         [ Links ]

21. Lee E, Schiller LR, Vendrell D, Santa Ana CA, Fordtran JS. Subepithelial collagen table thickness in colon specimens from patients with microscopic colitis and collagenous colitis. Gastroenterology 1992;103:1790-6.         [ Links ]

22. Munch A, Sodeholm JD, Wallon C, Ost A, Olaison G. Strom M. Dynamics of mucosal permeability and inflammation in collagenous colitis before, during and after loop ileostomy. Gut 2005;54:1126-8.         [ Links ]

23. Bohr J, Jarnerot G, Tysk C, Jones I, Eriksson S. Effect of fasting on diarrhoea in collagenous colitis. Digestion 2002;65:30-4.         [ Links ]

24. Bürgel N, Bojarski C, Mankertz J, Zeitz M, Fromm M, Schulzke JD. Mechanisms of diarrhoea in collagenous colitis. Gastroenterology 2002;123:433-43.         [ Links ]

25. Protic M, Jojic N, Bojic D, Milutinovic S, Necic D, Bojic B, et al. Mechanism of diarrhea in microscopic colitis. World J Gastroenterol 2005;11:5535-9.         [ Links ]

26. Walter SA, Munch A, Ost A, Strom M. Anorectal function in patients with collagenous colitis in active and clinically quiescent phase, in comparision with healthy controls. Neurogastroenterol Motil 2010;22:534-8.         [ Links ]

27. Erim T, Alazmi WM, O'Loughlin CJ, Barkin JS. Collagenous colitis associated with Clostridium difficile: A cause effect? Dig Dis Sci 2003;48:1374-5.         [ Links ]

28. Bohr J, Nordfelth R, Jarnerot G, Tysk C. Yersinia species in collagenous colitis: A serologic study. Scand J Gastroenterol 2002;37:711-14.         [ Links ]

29. Spiller R, Garsed K. Postinfectious irritable bowel syndrome. Gastroenterology 2009;136:1979-88.         [ Links ]

30. El-Salhy M, Gundersen D, Hatlebakk JG, Hausken T. Clinical presentation, diagnosis, pathogenesis and treatment options for lymphocytic colitis. Int J Molec Med 2013;32:263-70.         [ Links ]

31. Breuer NF, Rampton DS, Tammar A, Murphy GM, Dowling RH. Effect of colonic perfusion with sulfated and nonsulfated bile acids on mucosal structure and function in the rat. Gastroenterology 1983;84:969-77.         [ Links ]

32. Lewis FW, Warren GH, Goff JS. Collagenous colitis with involvement of terminal ileum. Dig Dis Sci 1991;36:1161-63.         [ Links ]

33. Ingle SB, Adgaonkar BD, Hinge CR. Microscopic colitis: Common cause of unexplained nonbloody diarrhea. World J Gastroenterol 2014;5:48-53.         [ Links ]

34. Fernandez-Bañares F, Esteve M, Salas A, Forné TM, Espinos JC, Martin-Comin J, et al. Bile acid malabsorption in microscopic colitis and in previously unexplained functional chronic diarrhea. Dig Dis Sci 2001;46:2231-8.         [ Links ]

35. Fausa O, Foerster A, Hoving T. Collagenous oclitis. A clinical, histological and ultrastructural study. Scan J Gastroenterol Suppl 1985;107:561-4.         [ Links ]

36. Kingham JG, Levison DA, Ball JA, Dawson AM. Microscopic colitis-a cause of chronic watery diarrhea. Br Med J (Clin Res Ed) 1982;107:8-23.         [ Links ]

37. Giardiello FM, Bayless TM, Jessurun J, Hamilton SR, Yardely JH. Collagenous colitis: Physiologic and histophatologic studies in seven patients. Ann Intern Med 1987;106:46-9.         [ Links ]

38. Laing AW, Pardi DS, Loftus EV Jr, Smyrk TC, Kammer PP, Tremaine WJ, et al. Microscopic colitis is not associated with cholecystectomy or appendectomy. Inflamm Bowel Dis 2006;12:708-11.         [ Links ]

39. Bohr J, Tysk C, Eriksson S, Abrahamsson H, Järnerot G. Collagenous colitis: A retrospecive study of clinical presentation and treatment in 163 patients. Gut 1996;39:846-51.         [ Links ]

40. Münch A, Soderholm JD, Ost A, Carlsson AH, Magnusson KE, Storm M. Low levels of bile acids increase bacterial uptake in colonic biopsies from patients with collagenous colitis in remission. Alimen Pharmacol ther 2011;33:954-60.         [ Links ]

41. Ung KA, Gillberg R, Kilander A, Abrahamsson H. role of bile acids and bile acid binding agents in patients with collagenous colitis. Gut 2000;46:170-5.         [ Links ]

42. Beaugerie L, Pardi DS. Review article: drug-induced microscopic colitis - proposal for a scoring system and review of the literature. Aliment Pharmacol Ther 2005;22:277-84.         [ Links ]

43. Bégaud B, Evreux JC, Jouglard J, Lagier G. Unexpected or toxic drug reaction assessment (imputation). Actualization of the method used in France. Therapie 1985;40:111-8.         [ Links ]

44. Piche T, Raimondi V, Schneider S, Hébuterne X, Rampal P. Acarbose and lymphocytic colitis. Lancet 2000;356:1246.         [ Links ]

45. Beaugerie L, Luboinski J, Brousse N, Cosnes J, Chatelet FP, Gendre FP, et al. Drug induced lymphocytic colitis. Gut 1994; 35: 426-8.         [ Links ]

46. Wilcox GM, Mattia A. Collagnous colitis associated with lansoprazole. J Clin Gastroenterol 2002;34:164-6.         [ Links ]

47. Wilcox G, Mattia AR. Microscopic colitis associated with omeprazole and esomeprazole exposure. J Clin Gastroenterol 2009;43:551-3.         [ Links ]

48. Beaugerie L, Patey N, Brousse N. Ranitidine, diarrhea and lymphocytic colitis. Gut 1995;37:708-11.         [ Links ]

49. Chassany O, Michaux A, Bergmann JF. Drug-induced diarrhoea. Drug Saf 2000;22:53-72.         [ Links ]

50. John LJ, Fromm M, Schulzke JD. Epithelial barriers in intestinal inflammation. Antioxid Redox Signal 2011;15:1255-70.         [ Links ]

51. Wildt S, Madsen JL, Rumessen JJ. Small-bowel permeability in collagenous colitis. Scand J Gastroenterol 2006;41:1044-49.         [ Links ]

52. Munch A, Soderholm JD, Ost A, Strom M. Increased transmucosal uptake of E. coli K12 in collagenous colitis persists after budesonide treatment. Am J Gastroenterol 2009;104:679-85.         [ Links ]

53. Munch A, Soderholm JD, Wallon C, Ost A, Olaison G, Strom M. Dynamics of mucosal permeability and inflammation in collagenous colitis before, during and after loop ileostomy. Gut 2005;54:1126-28.         [ Links ]

54. Pemer A, Nordgaard I, Matzen P, Rask-Madsen J. Colonic production of nitric oxide gas in ulcerative colitis, collagenous colitis and uninflamed bowel. Scand J Gastroenterol 2002;37:183-8.         [ Links ]

55. Olesen M, Middelveld R, Bohr J, Tysk C, Lundberg JO, Eriksson S, et al. Luminal nitric oxide and epithelial expression of inducible and endothelial nitric oxide synthase in collagenous and lymphocytic colitis. Scand J Gastroenterol 2003;38:66-72.         [ Links ]

56. Menconi MJ, Unno N, Smith M, Aguirre DE, Fink MP. Nitric oxide donor-induced hyperpermeability of cultured intestinal epithelial monolayers: Role of superoxide radical, hydroxyl radical and peroxynitrite. Biochim Biophys Acta 1998;1425:189-203.         [ Links ]

57. El-Salhy M, Gundersen D, Hatlebakk JG, Hausken T. Clinical presentation, diagnosis, pathogenesis and treatment options for lymphocytic colitis. Int J Molecular Med 2013;32:263-70.         [ Links ]

58. El-Salhy M, Gundersen D, Hatlebakk JG, Hausken T. High densities of serotonin and peptide YY cells in the colon of patients with lymphocytic colitis. World J Gastroenterol 2012;18:6070-75.         [ Links ]

59. Goumain M, Voisin T, Lorinet AM, Ducroc R, Tsocas A, Rozé C, et al. The peptide YY-preferring receptor mediating inhibition of small intestinal secretion is a peripheral Y(2) receptor: Pharmacological evidence and molecular cloning. Mol Pharmacol 2001;60:124-34.         [ Links ]

60. Souli A, Chariot J, Voisin T, Presset O, Tsocas A, Balasubramaniam A, et al. Several receptors mediate the antisecretory effect of peptide YY, neuropeptide Y and pancreatic polypeptide on VIP-induced fluid secretion in the rat jejunum in vivo. Peptides 1997;18:551-7.         [ Links ]

61. Whang EE, Hines OJ, Reeve JR, Grandt D, Moser JA, Bilchik AJ, et al. Antisecretory mechanisms of peptide YY in rat distal colon. Dig Dis Sci 1997;42:1121-7.         [ Links ]

62. Kumawat AK, Elgbratt K, Tysk C, Bohr J, Hörnquist EH. Reduced T cell receptor excision circle levels in the colonic mucosa of microscopic colitis patients indicate local proliferation rather than homing of peripheral lymphocytes to the inflamed mucosa. BioMed Res Int 2013;408638.         [ Links ]

63. Mosnier JF, Larvol L, Barge J, Dubois S, De La Bigne G, Hénin D, et al. Lymphocytic and collagenous colitis: An immunohistochemical study. Am J Gastroenterol 1996;91:709-13.         [ Links ]

64. Kumawat A, Strid H, Tysk C, Bohr J, Hörnquist EH. Microscopic colitis patients demonstrate a mixed Th17/Tc17 and Th1/Tc1 mucosal cytokine profile. Mol Immunol 2013;55:355-64.         [ Links ]

65. Kumawat AK, Strid H, Elgbratt K, Tysk C, Bohr J, Hultgren Hörnquist E. Microscopic colitis patients have increased proportions of Ki67+ proliferating and CD405RO+ active/memory CD8+ and CD4+8+ mucosal T cells. J Crohns Colitis 2013;7:694-705.         [ Links ]

66. Ablamunits V, Bisikirska BC, Herold KC. Human regulatory CD8T cells. Ann N Y Acad Sci 2008;1150:234-8.         [ Links ]

67. Shalev I, Schmelzle M, Robson SC, Levy G. Making sense of regulatory T cell suppressive function. Semin Immunol 2011;23:282-92.         [ Links ]

68. Trzonkowski P, Szmit E, Mysliwska J, Mysliwski A. CD4+CD25+ T regulatory cells inhibit cytotoxic activity of CTL and NK cells in humans-impact of immunosenescence. Clin Immunol 2006;119:307-16.         [ Links ]

69. Maloy KJ, Powrie F. Regulatory T cells in the control of immune pathology. Nat Immunol 2001;2:816-22.         [ Links ]

70. Shuting Bai, Gene PS, Nirag CJ. Foxp3 expression patterns in microscopic colitis. Am J Clin Pathol 2012;137:931-6.         [ Links ]

71. Fernandez-Bañares F, Casalots J, Salas A, Esteve M, Rosinach M, Forné M, et al. Paucicellular lymphocytic colitis: is it a minor form of lymphocytic colitis? A clinical pathological and immunological study. Am J Gastroenterol 2009;104:1189-98.         [ Links ]

72. Gunther U, Bateman AC, Beattie RM, Bauer M, MacDonald TT, Kaskas BA. Connective tissue growth factor expression is increased in collagenous colitis and coeliac disease. Histopathology 2010;57:427-35.         [ Links ]

73. Wildgren S, Jlidi R, Cox JN. Collagenous colitis: Histologic, morphometric, immunohistochemical and ultrastructural studies. Report of 21 cases. Virchows Arch A Pathol Anat Histopathol 1988;413:287-96.         [ Links ]

74. Salas A, Fernandez-Bañares F, Casalots J, González C, Tarroch X, Forcada P, et al. Subepithelial myofibroblasts and tenascin expression in microscopic colitis. Histopathology 2003;43:48-54.         [ Links ]

75. Griga T, Tromm A, Schmiegel W, Pfisterer O, Muller KM, Brasch F. Collagenous colitis: Implications for the role of vascular endothelial growth factor in repair mechanisms. Eur J Gastroenterol Hepatol 2004;16:397-402.         [ Links ]

76. Wagner M, Lampinen M, Sangfelt P, Agnarsdottir M, Carison M. Budesonide treatment of patients with collagenous colitis restores normal eosinophil and T-cell activity in the colon. Inflamm Bowel Dis 2010;16:1522-8.         [ Links ]

77. Levy AM, Yamazaki K, Van Keulen VP, Burgart LJ, Sandborn WJ, Philips SF, et al. Increased eosinophil infiltration and degranulation in colonic tissue form patients with collagenous colitis. Am J Gastroenterol 2001;96:1522-8.         [ Links ]

78. Taha Y, Carison M, Thorn M, Loof L, Raab Y. Evidence of local eosinophil activation and altered mucosal permeability in collagenous colitis. Dig Dis Sci 2001;46:888-97.         [ Links ]

79. Dey I, Beck PL, Chadee K. Lymphocytic colitis is associated with increased pro-inflammatory cytokine profile and up regulation of prostaglandin receptor EP4. PloS One 2013;8:e61891.         [ Links ]

80. Andersen I, Jorgensen VL, Perner A, Eugen-Olsen J, Rask-Madsen J. Activation of nuclear factor KappaB in colonic mucosa from patients with collagenous colitis and ulcerative colitis. Gut 2005; 54:503-9.         [ Links ]

81. Tagkalidis PP, Gibson PR, Bhathal PS. Microscopic colitis demonstrates a T helper cell type 1 mucosal cytokine profile. J Clin Pathol 2007;60:382-7.         [ Links ]

82. Perner A, Andersen L, Normark M, Fischer-Hansen B, Sorensen S, Eugen-Olsen J, et al. Expression of nitric oxide synthases and effects of L-arginine and L-NMMA on nitric oxide production and fluid transport in collagenous colitis. Gut 2001;49:387-94.         [ Links ]

83. Wildt S, Rumessen JJ, Csillag C, Normark M, Poulsen KA, Kolko M. Cycloxygenase-2 immunoreactivity in collagenous colitis. APMIS 2009;117:500-6.         [ Links ]

84. Park E, Park YS, Park DR, Jung SA, Han DS, Jang BI, et al. Cytokine expression of microscopic colitis including interleukin-17. Gut Liver 2014; DOI: 10.5009/gnl13439.         [ Links ]

85. Järnerot G, Hertervig E, Grännö C, Thorhallsson E, Eriksson S, Tysk C, et al. Familial occurrence of microscopic colitis: a report on five families. Scand J Gastroenterol 2001;36:959-62.         [ Links ]

86. Van Tilburg AJ, Lam HG, Seldenrijk CA, Stel HV, Block P, Dekker W, et al. Familial occurrence of collagenous colitis. A report of two families. J Clin Gastroenterol 1990;12:279-85.         [ Links ]

87. Thomson A, Kaye G. Further report of familial occurrence of collagenous colitis. Scand J Gastroenterol 2002;37:1244-6.         [ Links ]

88. Phull PS, Vijayan B, Bisset WM, Murray GI. Familial collagenous colitis involving a 6-year old child. J Crohn's Colitis 2012;6:606-9.         [ Links ]

89. Giardiello FM, Lazenby AJ, Yardeley JH, Bias WB, Johnson J, Alianiello RG, et al, Increased HLA A1 and diminished HLA A3 in lymphocytic colitis compared to controls and patients with collagenous colitis. Dig Dis Sci 1992;37:496-9.         [ Links ]

90. Fine KD, Do K, Schulte K, Ogunji F, Guerra R, Osowski L, et al. High prevalence of celiac sprue-like HLA-DQ genes and enteropathy in patients with the microscopic colitis syndrome. Am J Gastroenterol 2000;95:1974-82.         [ Links ]

91. Fernandez-Bañares F, Esteve M, Farré C, Salas A, Alsina M, Casalots J, et al. Predisposing HLA-DQ2 and HLA-DQ8 haplotypes of coeliac disease and associated enteropathy in microscopic colitis. Eur J Gastroenterol Hepatol 2005;17:1333-8.         [ Links ]

92. Madisch A, Hellmig S, Schreiber S, Bethke B, Stolte M, Miehlke S. Allelic variation of the matrix metalloproteinase-9 gene is associated with collagenous colitis. Inflamm Bowel Dis 2011;17:2295-8.         [ Links ]

93. Castaneda FE, Walia B, Vijay-Kumar M, Patel NR, Roser S, Kolachala VL, et al. Targeted deletion of metalloproteinase 9 attenuates experimental colitis in mice: Central role of epithelial-derived MMP. Gastroenterology 2005;129:1991-2008.         [ Links ]

94. Koskela RM, Karttunen RA, Niemelä SE, Lehtola JK, Bloigu RS, Karttunen RA. Cytokine gene polymorphism in microscopic colitis association with the IL-6-174 GG genotype. Eur J Gastroenterol Hpeatol 2011;23:607-13.         [ Links ]

95. Koskela RM, Karttunen TJ, Niemelá SE, Lehtola JK, Llonen J, Karttunen RA. Human leucocyte antigen and TNFα polymorphism association in microscopic colitis. Eur J Gastroenterol Hepatol 2008;20:276-82.         [ Links ]

96. Baert F, Schmit A, D'Haens G, Dedeurwaerdere F, Louis E, Cabooter M, et al. Budesonide in collagenous colitis: A double-blind placebo-controlled trial with histologic follow-up. Gastroenterology 2002;122:20-5.         [ Links ]

97. Bonderup OK, Hansen JB, Birket-Smith L, Vestergaard V, Teglbjaerg PS, Fallingborg J. Budesonide treatment of collagenous colitis: A randomised, double blind, placebo controlled trial with morphometric analysis. Gut 2003;52:248-51.         [ Links ]

98. Miehlke S, Heymer P, Bethke B, Bastlein E, Meier E, Bartram HP, et al. Budesonide treatment for collagenous colitis: A randomized, double-blind, placebo-controlled, multicenter trial. Gastroenterology 2002;123:978-84.         [ Links ]

99. Miehlke S, Madisch A, Karimi D, Wonschik S, Kuhlisch E, Beckmann R, et al. Budesonide is effective in treating lymphocytic colitis: A randomized double-blind placebo-controlled study. Gastroenterology 2009;136:2092-100.         [ Links ]

100. Pardi DS, Loftus Jr EV, Tremaine WJ, Sandborn WJ. A randomized double-blind, placebo controlled trial of budesonide for the treatment of active lymphocytic colitis. Gastroenterology 2009;136(Supl. 1)(T 1193).         [ Links ]

101. Miehlke S, Madisch A, Kupcinskas L, Petrauskas D, Bohm G, Marks HJ, et al. Budesonide is more effective than mesalamine or placebo in short-term treatment of collagenous colitis. Gastroenterology 2014;146:1222-30.         [ Links ]

102. Chande N, MacDonald JK, McDonald JW. Interventions for treating microscopic colitis: A Cochrane inflammatory bowel disease and functional bowel disorders review group systematic review of randomized trials. Am J Gastroenterol 2009; 104:235-41.         [ Links ]

103. Chande N, McDonald JW, MacDonald JK. Interventions for treating lymphocytic colitis. Cochrane Database Syst Rev 2008 (CD006096)).         [ Links ]

104. Stewart MJ, Seow CH, Storr MA. Prednisolone and budesonide for short- and long-term treatment of microscopic colitis: Systematic review and meta-analysis. Clin Gastroenterol Hepatol 2011;9:881-90.         [ Links ]

105. Bonderup OK, Hansen JB, Teglbjaerg PS, Christensen LA, Fallingborg JF. Long-term budesonide treatment of collagenous colitis: A randomised, double-blind, placebo-controlled trial. Gut 2009;58:68-72.         [ Links ]

106. Miehlke S, Madisch A, Bethke B, Morgner A, Kuhlisch E, Henker C, et al. Oral budesonide for maintenance treatment of collagenous colitis: A randomized, double-blind, placebo-controlled trial. Gastroenterology 2008;135:1510-6.         [ Links ]

107. Münch A, Bohr J, Miehlke S, Benoni C, Olesen M, Ost A, et al. Low-dose budesonide for maintenance of clinical remission in collagenous colitis: A randomised, placebo-controlled, 12 month trial. Gut 2014; DOI: 10.1136/gutjnl-2014-308363.         [ Links ]

108. McKay DM, Brattsand R, Wieslander E, Fung M, Croitoru K, Perdue MH. Budesonide inhibits T cell-initiated epithelial pathophysiology in an in vitro model of inflammation. J Pharmacol Exp Ther 1996;277:403-10.         [ Links ]

109. Göke MN, Schneider M, Beil W, Manns MP. Differential glucocorticoid effects on repair mechanisms and NF-kappaB activity in the intestinal epithelium. Regul Pept 2002;30:203-14.         [ Links ]

110. Barnes PJ. Molecular mechanisms and cellular effects of glucocorticosteroids. Immunol Allergy Clin North Am 2005;25:451-68.         [ Links ]

111. Bloemena E, Weinreich S, Schellekens PT. The influence of prednisolone on the recirculation of peripheral blood lymphocytes in vivo. Clin Exp Immunol 1990;80:460-6.         [ Links ]

112. Wagner M, Lampinen M, Sangfelt P, Agnarsdottir M, Carison M. Budesonide treatment of patients with collagenous colitis restores normal eosinophil and T-cell activity in the colon. Inflamm Bowel Dis 2010;16:1118-26.         [ Links ]

113. Baert F, Schmit A, D'Haens G, Dedeurwaerdere F, Louis E, Cabooter M, et al. Budesonide in collagenous colitis: A double-blind placebo-controlled trial with histologic follow-up. Gastroenterology 2002;122:20-5.         [ Links ]

114. Bonderup OK, Hansen JB, Madsen P, Vestergaard V, Fallingborg J, Teglbjaerg PS. Budesonide treatment and expression of inducible nitric oxide synthase mRNA in colonic mucosa in collagenous colitis. Eur J Gastroenterol Hepatol 2006;18:1095-9.         [ Links ]

115. Bajor A, Kilander A, Galman C, Rudling M, Ung KA. Budesonide treatment is associated with increased bile acid absorption in collagenous colitis. Aliment Pharmacol Ther 2006;24:1643-9.         [ Links ]

116. Barmeyer C, Erko I, Fromm A, Bojarski C, Allers K, Moos V, et al. Ion transport and barrier function are disturbed in microscopic colitis. Ann N Y Acad Sci 2012;1258:143-8.         [ Links ]

117. Olesen M, Eriksson S, Bohr J, Jarnerot G, Tysk C. Lymphocytic colitis: A retrospective clinic study of 199 Swedish patients. Gut 2004;53:536-41.         [ Links ]

118. Bohr J, Tysk C, Eriksson S, Abrahamsson H, Järnerot G. Collagenous colitis: A retrospective study of clinical presentation and treatment in 163 patients. Gut 1996;39:846-51.         [ Links ]

119. Fernández-Bañares F, Salas A, Esteve M, Espinós J, Forné M, Viver JM. Collagenous colitis and lymphocytic colitis. Evaluation of clinical and histological features, response to treatment and long-term follow-up. Am J Gastroenterol 2003;98:340-7.         [ Links ]

120. Pardi DS, Ramnath VR, Loftus EV, Tremaine WJ, Sandborn WJ. Lymphocytic colitis: Clinical features, treatment, and outcomes. Am J Gastroenterol 2002;97:2829-33.         [ Links ]

121. Calabrese C, Fabbri A, Areni A, Zahlane D, Scialpi C, Di Febo G. Mesalazine with or without cholestyramine in the treatment of microscopic colitis: Randomized controlled trial. J Gastroenterol Hepatol 2007;22:809-14.         [ Links ]

122. Ung KA, Gillberg R, Kilander A, Abrahamsson H. role of bile acids and bile acid binding agents in patients with collagenous colitis. Gut 2000;46:170-5.         [ Links ]

123. Ung KA, Kilander A, Nilsson O, Abrahamsson H. Long-term course in collagenous colitis and the impact of bile acids malabsorption and bile acid sequestrants on histopathology and clinical features. Scand J Gastroenterol 2001;36:601-9.         [ Links ]

124. Park YS, Baek DH, Kim WH, Kim JS, Yang SK, Jung SA, et al. Clinical characteristics of microscopic colitis in Korea: Prospective multicenter study by KASID. Gut Liver 2011;5:181-6.         [ Links ]

125. Wildt S, Munck LK, Vinter-Jensen L, Hanse BF, Nordgaard-Lassen I, Christesen S, et al. Probiotic treatment of collagenous colitis: A randomized, double-blind placebo-controlled trial with Lactobacillus acidophilus and Bifidobacterium animalis subsp. Lactis. Inflamm Bowel Dis 2006;12:395-401.         [ Links ]

126. Madisch A, Miehlke S, Eichele O, Mrwa J, Bethke B, Kuhlisch E, et al. Boswellia serrata extract for the treatment of collagenous colitis. A double-blind, randomized, placebo-controlled, multicenter trial. Int J Colorectal Dis 2007;22:1445-51.         [ Links ]

127. Tromm A, Niewerth U, Khoury M, Baestlein E, Wilhelms G, Schulze J, et al. The probiotic E. coli strain Nissle 1917 for the treatment of collagenous colitis: First results of an open-label trial. Z gastroenterol 2004;42:365-9.         [ Links ]

128. Fine KD, Ogunji F, Lee EL, Lafon G, Tanzi M. Randomized, double-blind, placebo-controlled trial of bismuth subsalicylate for microscopic colitis. Gastroenterology 1999;116:A880.         [ Links ]

129. Fine KD, Lee EL. Efficacy of open-label bismuth subsalicylate for the treatment of microscopic colitis. Gastroenterology 1998;114:29-36.         [ Links ]

130. Gentile NM, Abdalla AA, Khanna S, Smyrk TC, Tremaine WJ, Faubion WA, et al. Outcomes of patients with microscopic colitis treated with corticosteroids: A population-based study. Am J Gastroenterology 2013;108:256-9.         [ Links ]

131. Pardi DS, Loftus EV, Tremaine WJ, Sandborn WJ. Treatment of refractory microscopic colitis with azathioprine and 6-mercaptopurine. Gastroenterology 2001;120:1483-4.         [ Links ]

132. Vennamaneni SR, Bonner GF. Use of azathioprine or 6-mercaptopurine for treatment of steroid-dependent lymphocytic and collagenous colitis. Am J Gastroenterol 2001;96:2798-9.         [ Links ]

133. Riddell J, Hillman L, Chiragakis L, Clarke A. Collagenous colitis: oral low-dose methotrexate for patients with difficult symptoms: Long term outcomes. J Gastroenterol Hepatol 2007;22:1589-93.         [ Links ]

134. Münch A, Bohr J, Vigren L, Tysk C, Ström M. Methotrexate is not effective in budesonide refractory collagenous colitis. Gut 2011;60(Supl. 3):A406.         [ Links ]

135. Esteve M, Mahadevan U, Sainz E, Rodríguez E, Salas A, Fernández-Bañares F. Efficacy of anti-TNF therapies in refractory severe microscopic colitis. J Crohns Colitis 2011;5:612-8.         [ Links ]

136. Münch A, Ignatova S, Strom M. Adalimumab in budesonide and methotrexate refractory collagenous colitis. Scand J Gastroenterol 2012;47:59-63.         [ Links ]

137. Dey I, Beck PL, Chadee K. Lymphocytic colitis is associated with increased pro-inflammatory cytokine profile and up regulation of prostaglandin receptor EP4. PLoS One 2013;8:e61891.         [ Links ]

138. Kumawat AK, Strid H, Tysk C, Bohr J, Hörnquist EH. Microscopic colitis patients demonstrate a mixed Th17/Tc17 and Th1/Tc1 mucosal cytokine profile. Mol Immunol 2013;55:355-64.         [ Links ]

139. Zhang Z, Zheng M, Bindas J, Schwarzenberger P, Kolls JK. Critical role of IL-17 receptor signaling in acute TNBS-induced colitis. Inflamm Bowel Dis 2006;12:382-8.         [ Links ]

 

 

Correspondence:
Danila Guagnozzi
Department of Gastroenterology.
Hospital Dr José
Molina Orosa.
Ctra. Arrecife-Tinajo, km. 1,300
35550 Arrecife, Las Palmas. Spain
e-mail: danila_g@libero.it

Received: 02-01-2015
Accepted: 08-01-2015

Creative Commons License Todo el contenido de esta revista, excepto dónde está identificado, está bajo una Licencia Creative Commons