Elsevier

Biochemical Pharmacology

Volume 98, Issue 3, 1 December 2015, Pages 422-431
Biochemical Pharmacology

Lack of TNFRI signaling enhances annexin A1 biological activity in intestinal inflammation

https://doi.org/10.1016/j.bcp.2015.09.009Get rights and content

Abstract

We evaluated whether the lack of TNF-α signaling increases mucosal levels of annexin A1 (AnxA1); the hypothesis stems from previous findings showing that TNF-α neutralization in Crohn’s disease patients up-regulates systemic AnxA1 expression. Biopsies from healthy volunteers and patients under anti-TNF-α therapy with remittent ulcerative colitis (UC) showed higher AnxA1 expression than those with active disease. We also evaluated dextran sulfate sodium (DSS)-acute colitis in TNF-α receptor 1 KO (TNFR1−/−) strain with impaired TNF-α signaling and C57BL/6 (WT) mice. Although both strains developed colitis, TNFR1−/− mice showed early clinical recovery, lower myeloperoxidase (MPO) activity and milder histopathological alterations. Colonic epithelium from control and DSS-treated TNFR1−/− mice showed intense AnxA1 expression and AnxA1+ CD4+ and CD8+ T cells were more frequent in TNFR1−/− animals, suggesting an extra supply of AnxA1. The pan antagonist of AnxA1 receptors exacerbated the colitis outcome in TNFR1−/− mice, supporting the pivotal role of AnxA1 in the early recovery. Our findings demonstrate that the TNF-α signaling reduction favors the expression and biological activity of AnxA1 in inflamed intestinal mucosa.

Introduction

Annexin A1 (AnxA1) is a phospholipid binding protein able to inhibit leukocyte transmigration and activation. Originally described as a glucocorticoid (GC)-induced factor with anti-phospholipase activity, later on AnxA1 has been implicated in prevention of inflammation in several disease models, such as peritonitis, air-pouch edema, myocardium infarct, respiratory tract allergy, endotoxemia, kidney ischemia/reperfusion, uveitis, arthritis and others [1], [2], [3], [4], [5], [6], [7], [8], [9]. AnxA1 binds to formyl peptide receptors (FPR) [10] and supports the resolution of inflammation and the wound healing suggesting a protective role on the intestinal epithelium damage [11], [12], [13], [14].

Inflammatory bowel diseases (IBD) are chronic gut illnesses. Between them, ulcerative colitis (UC) is a prevailing entity with continuous superficial colonic inflammation, abdominal pain and recurrent diarrhea [15]. The proposed mechanism for UC development is the breakdown of the interaction between environmental stimuli and gut microbiota that results in mucosal inflammation [16]. This change may include disruption of the barrier function and alteration in the regulation of acute and adaptive immune responses [17].

The pro-inflammatory cytokine TNF-α plays a pivotal role in the signaling cascade that causes chronic intestinal inflammation in IBD [18]. Biologic agents such as anti-TNF-α antibodies Infliximab (IFX) and Adalimumab (ADA) attenuate the inflammatory process and have been approved for the treatment of moderate-to-severe UC that is refractory to conventional therapy [19]. These antibodies are well tolerated, induce and maintain clinical remission and mucosal healing, and permit the tapering of corticosteroids while allow remission [19], [20]. Interestingly, the anti-TNF-α therapy produces early changes in the gene expression profiles of intestinal epithelial cells that could be predictive of clinical responses [21], [22]. The availability of predictive correlates of clinical response is highly relevant as could enable to determine the benefits or risks of enduring biologic therapies on individual basis. In agreement, in a longitudinal assessment of peripheral blood samples from IBD patients recently we showed that systemic mononuclear cell transcripts as well as plasma levels of the anti-inflammatory biomarker AnxA1 are affected by initial and continuous IFX therapy [23]. Surprisingly, the response correlated with lower C-reactive protein and better quality of life of these patients.

We hypothesized that the absence or reduction of TNF-α signaling could amplify mucosal levels of the protein AnxA1. Herein, biopsies from patients under anti-TNF-α therapy during UC remission showed an increase in mucosal AnxA1 expression. We also used mice lacking TNFR1 (transmembrane and soluble receptor of TNF-α) to mimic the activity of anti TNF-α antibody, and we examined the expression and anti-inflammatory activity of AnxA1 in the colon mucosa in the well characterized animal model of dextran sulfate sodium (DSS) induced colitis. We found that the blockade of the TNF-α pathway upregulated the AnxA1 expression in the epithelium and colon mucosa and simultaneously accelerated the resolution process of the colitis.

Section snippets

Ethic statement

This study was approved by the Ethics Committee of the Hospital Privado-Centro Medico de Córdoba (approval no. HP 4-215). All patients medically assisted in the 2013 year gave their written informed consent prior to participation in this study. Animal experiments were approved by and conducted in accordance with guidelines of the Committee for Animal Care and Use of the Chemical Science Faculty, National University of Cordoba (approval no. HCD 15-09-69596) in strict accordance with the

AnxA1 expression in colon from UC patients in remission under anti TNF-α therapy

Previously, IFX treated Crohn’s patients presented increase of AnxA1 plasma levels and up-regulation of AnxA1 mRNA transcription in peripheral mononuclear cells [23]. We wondered if biological agents such as IFX could also modify levels of AnxA1 in colonic mucosa in IBD patients. To address this question we explored the expression and localization of AnxA1 in biopsy sections of UC patients either untreated or in remission upon anti-TNF-α therapy. For comparative purposes we also included UC

Discussion

Many reports support the intimate relationship of TNFR1 expression and signaling with the pathogenesis of inflammatory diseases. Some studies have explored the presence of protein biomarkers in serum/plasma of patients with rheumatoid arthritis treated with IFX, one of the major anti-TNF-α drugs [33], [34]. Mainly, up- or down-differentially regulated proteins were apolipoproteins, components of the complement system and acute phase reactants [33], [34].

Herein we found that patients who were

Conflict of interest

The authors declare no conflict of interest.

Acknowledgments

Dextran Sulphate 40 000 MW was a kind gift of George Usher, Dextran Products Limited (Canada). We would like to thank Paula Icely and Fabricio Navarro for excellent technical assistance. This work was supported by grants of Agencia Nacional de Promoción Científica y Tecnológica (FONCYT), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) and Secretaria de Ciencia y Tecnología (SECyT-UNC). SGC belongs to the CONICET research staff.

References (48)

  • A.S. Damazo et al.

    Endogenous annexin A1 counter-regulates bleomycin-induced lung fibrosis

    BMC Immunol.

    (2011)
  • F.N. Facio et al.

    Annexin 1 mimetic peptide protects against renal ischemia/reperfusion injury in rats

    J. Mol. Med. (Berl.)

    (2011)
  • H.B. Patel et al.

    The impact of endogenous annexin A1 on glucocorticoid control of inflammatory arthritis

    Ann. Rheum. Dis.

    (2012)
  • A.P. Girol et al.

    Anti-inflammatory mechanisms of the annexin A1 protein and its mimetic peptide Ac2-26 in models of ocular inflammation in vivo and in vitro

    J. Immunol.

    (2013)
  • S. Ernst et al.

    An annexin 1 N-terminal peptide activates leukocytes by triggering different members of the formyl peptide receptor family

    J. Immunol.

    (2004)
  • B.A. Babbin et al.

    Annexin A1 regulates intestinal mucosal injury, inflammation, and repair

    J. Immunol.

    (2008)
  • N. Ouyang et al.

    MC-12, an annexin A1-based peptide, is effective in the treatment of experimental colitis

    PLoS One

    (2012)
  • L. Vong et al.

    Up-regulation of annexin-A1 and lipoxin A(4) in individuals with ulcerative colitis may promote mucosal homeostasis

    PLoS One

    (2012)
  • G. Leoni et al.

    Annexin A1, formyl peptide receptor, and NOX1 orchestrate epithelial repair

    J. Clin. Investig.

    (2013)
  • J. Cosnes et al.

    Epidemiology and natural history of inflammatory bowel diseases

    Gastroenterology

    (2011)
  • K.J. Maloy et al.

    Intestinal homeostasis and its breakdown in inflammatory bowel disease

    Nature

    (2011)
  • B. Khor et al.

    Genetics and pathogenesis of inflammatory bowel disease

    Nature

    (2011)
  • I. Ordas et al.

    Anti-TNF monoclonal antibodies in inflammatory bowel disease: pharmacokinetics-based dosing paradigms

    Clin. Pharmacol. Ther.

    (2012)
  • M. Bosani et al.

    Biologic targeting in the treatment of inflammatory bowel diseases

    Biologics

    (2009)
  • Cited by (20)

    • Soy genistein administered in soluble chitosan microcapsules maintains antioxidant activity and limits intestinal inflammation

      2018, Journal of Nutritional Biochemistry
      Citation Excerpt :

      On day 10, animals were sacrificed and colon removed for further studies. Colon segments were homogenized as previously [30,31]; supernatants were stored at −70 °C for further analysis. Murine TNF-α and MCP-1 (BD Biosciences), IL-6 and IL-10 (BD Pharmingen) were evaluated with ELISA kits as specified by the manufacturers.

    • Development of an XBP1 agonist, HLJ2, as a potential therapeutic agent for ulcerative colitis

      2017, European Journal of Pharmaceutical Sciences
      Citation Excerpt :

      The proinflammatory cytokines TNF-α, IL-1β and IL-6 play important roles in UC progression by recruiting and activating leukocytes in colon lesions (Yamamoto et al., 2006; Andoh et al., 2004); their levels can also be used as markers of disease severity in UC models. Likewise, MPO activity is regarded as an indicator of leukocyte function and activity and tracks with UC severity (Sena et al., 2015). While increased inflammatory cytokine levels and MPO activity were detected in tissue samples from mice with UC, HLJ2 treatment significantly diminished both indicators in a dose dependent manner.

    View all citing articles on Scopus
    View full text