Lactoferrin differently modulates the inflammatory ... - Springer Link

2 downloads 0 Views 538KB Size Report
Apr 26, 2014 - Lactoferrin differently modulates the inflammatory response in epithelial models mimicking human inflammatory and infectious diseases.
Biometals (2014) 27:843–856 DOI 10.1007/s10534-014-9740-9

Lactoferrin differently modulates the inflammatory response in epithelial models mimicking human inflammatory and infectious diseases Alessandra Frioni • Maria Pia Conte • Antimo Cutone • Catia Longhi • Giovanni Musci • Maria Carmela Bonaccorsi di Patti • Tiziana Natalizi Massimiliano Marazzato • Maria Stefania Lepanto • Patrizia Puddu • Rosalba Paesano • Piera Valenti • Francesca Berlutti



Received: 30 March 2014 / Accepted: 13 April 2014 / Published online: 26 April 2014 Ó Springer Science+Business Media New York 2014

Abstract Conflicting data are reported on pro- or anti-inflammatory activity of bovine lactoferrin (bLf) in different cell models as phagocytes or epithelial cell lines infected by bacteria. Here we evaluated the bLf effect on epithelial models mimicking two human pathologies characterized by inflammation and infection with specific bacterial species. Primary bronchial epithelium from a cystic fibrosis (CF) patient and differentiated intestinal epithelial cells were infected with Pseudomonas aeruginosa LESB58 isolated from a CF patient and Adherent-Invasive Escherichia coli LF82 isolated from a Crohn’s disease patient. Surprisingly, bLf significantly reduced the intracellular bacterial survival, but differently modulated the inflammatory response. These data lead us to hypothesize that bLf differentially acts depending on the

epithelial model and infecting pathogen. To verify this hypothesis, we explored whether bLf could modulate ferroportin (Fpn), the only known cellular iron exporter from cells, that, by lowering the intracellular iron level, determines a non permissive environment for intracellular pathogens. Here, for the first time, we describe the bLf ability to up-regulate Fpn protein in infected epithelial models. Our data suggest that the mechanism underlying the bLf modulating activity on inflammatory response in epithelial cells is complex and the bLf involvement in modulating cellular iron homeostasis should be taken into account. Keywords Lactoferrin  Inflammation  Cystic fibrosis  Chron’s disease  Ferroportin  Infection

This paper is dedicated to the memory of our colleague, Dr. Patriza Puddu who recently passed away. A. Frioni  M. P. Conte  A. Cutone  C. Longhi  T. Natalizi  M. Marazzato  M. S. Lepanto  P. Valenti  F. Berlutti (&) Microbiology Section, Department of Public Health and Infectious Diseases, University Sapienza of Rome, P.le A. Moro 5, 00185 Rome, Italy e-mail: [email protected]

M. C. B. di Patti Department of Biochemical Sciences, Sapienza University, P.le A. Moro 5, 00185 Rome, Italy

G. Musci Department of Biosciences and Territory, University of Molise, c.da Fonte Lappone, Pesche, IS, Italy

R. Paesano Department of Women’s Health and Territorial Medicine, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy

P. Puddu Istituto Superiore di Sanita`, Viale Regina Elena, 299, 00161 Rome, Italy

123

844

Introduction Lactoferrin (Lf), a key factor in the host defense system, belongs to innate non specific immune system (Wiesner and Vilcinskas 2010). Differently to the most antimicrobial proteins or other molecules of the immune system, Lf acts as modulator of the inflammatory process ranging from inhibition to promotion of inflammation. Literature data are crammed with results showing contradictory effects of Lf on inflammatory processes in in vitro different cell models. Of course, these conflicting data makes it difficult to decipher the actual Lf mechanisms through which the molecule acts. However, a deep analysis of conflicting in vitro results reveals the influence of experimental conditions as the use of phagocytic or epithelial cell lines, the different type of infecting agents as bacteria or viruses, or the stimulation with pathogen-associated molecular patterns (PAMPS) as flagellin, toxin, peptidoglycan, lypopolisaccharide (LPS), and, finally, the use of Lf from different sources (Puddu et al. 2007; Hayworth et al. 2009; Latorre et al. 2012; Legrand 2012). As matter of the fact, Lf is able to induce IL-6 production both in murine peritoneal macrophages trough both a TLR-4 dependent and independent pathway (Curran et al. 2006; Puddu et al. 2007) and in freshly isolated blood monocytes (Puddu et al. 2011). Conversely, the ability of Lf to bind LPS limits the interaction of this PAMP with cellular or soluble receptors, thus reducing the intensity of LPS-mediated inflammatory response of phagocytic cells (Puddu et al. 2011; Latorre et al. 2012). Unlike phagocytes, the epithelial cells are more responsive to live bacteria than to their components as LPS (Cario et al. 2000; Vora et al. 2004). The Lf ability in modulating inflammatory response was detectable only when the cell lines were injured using a strong stimulus such as living microorganisms or toxins (Berlutti et al. 2006; Valenti et al. 2011; Legrand 2012; Komatsu et al. 2013). In these models, Lf exerts essentially an antiinflammatory activity thus reducing the inflammatoryrelated cell damage. Cystic fibrosis (CF) is an autosomal recessive disease caused by mutations within the cystic fibrosis transmembrane conductance regulator (CFTR) (Boucher 2002). In airway epithelia, mutated CFTR leads to perturbations in the regulation of several intracellular signaling pathways resulting in excessive

123

Biometals (2014) 27:843–856

production of NF-jB-dependent cytokines such as interleukin (IL)-1, IL-6 and IL-8 (Nichols et al. 2007). Moreover, the CFTR dysfunction leads to abnormalities in iron transport across the airway epithelial cells (O’Sullivan and Flume 2009) and to iron homeostasis disorders (Moreau-Marquis et al. 2008; Wang 2010) as demonstrated by the altered expression of the storage protein ferritin (Ft), the iron importer DMT1, and the exporter ferroportin (Fpn) (Ghio et al. 2005). The high iron concentrations in airway secretions (Reid et al. 2004, 2007) precede the introduction of microbes into the respiratory tract favoring their multiplication and contributing to an increased production of reactive oxygen species that in turn enhance the inflammatory status (Khan et al. 1995; Tirouvanziam et al. 2002). Therefore, the airway of CF subjects is inherently prone to infection that begins in very early life (Ranganathan et al. 2011). By adulthood, the chronic airway infection is sustained in the 80 % of cases by Pseudomonas aeruginosa (Tunney et al. 2008). The iron excess in CF airway secretions stimulates P. aeruginosa to adopt the biofilm lifestyle (Lee et al. 2003; Berlutti et al. 2005; Moreau-Marquis et al. 2008). Bacterial biofilm invades CF airway epithelial cells that respond by activating a strong inflammatory response through elevation of IL-6 and IL-1b levels that, in turn, induces cell damage (Moreau-Marquis et al. 2008; Sagel et al. 2009; Valenti et al. 2011). Crohn’s disease (CD) is a chronic inflammatory bowel disease, characterized by a deregulate mucosal immune response to gut microbiota. Immunologically, CD patients produce high amounts of cytokines including interferon (IFN)-c in the inflamed lamina propria as the result of abnormal Th1- and Th17mediated immune responses (Strober and Fuss 2011). Further evidences support the idea that adherentinvasive Escherichia coli (AIEC), a commensal with the potential to cause disease, may have a causative role in initiating and perpetuating the intestinal inflammation in CD patients (Sasaki et al. 2007; Rolhion and Darfeuille-Michaud 2007; MartinezMedina et al. 2009; Chassaing et al. 2013). AIEC strains colonize the ileal epithelium of CD patients and are able to adhere to and invade intestinal epithelial cells and to replicate within macrophages (Glasser et al. 2001; Darfeuille-Michaud et al. 2004). AIEC adhesion/invasion is dependent on the expression of both type 1 pili expression on the bacterial surface and

Biometals (2014) 27:843–856

of carcinoembryonic antigen–related cell adhesion molecule (CEACAM6) expression on the apical surface of intestinal epithelial cells. Moreover it has been demonstrated that CEACAM6 expression in cultured intestinal epithelial cells is upregulated both by AIEC infection and by stimulation with the proinflammatory cytokine IFN-c (Barnich et al. 2007). Lf levels have been shown to increase dramatically in body fluids during the course of inflammation. In particular, in CF bronchial secretions the high levels of pro-inflammatory cytokines, including IL-8, recruit neutrophils which synthesize and secrete Lf. As a consequence, Lf is found at higher concentrations in airway secretion of CF patients than in healthy humans (up to 0.1 and 0.01 mg/ml, respectively) (Sagel et al. 2009). In CD patients the high levels of Lf found in feces are proportional to the neutrophil flux (Pfefferkorn et al. 2010). Here we evaluate the role of bovine Lf (bLf) in modulating the inflammatory responses in two epithelial cell models: primary human airway epithelium deriving from CF patients and colonic epithelial cells. The epithelia cell models were infected by P. aeruginosa LESB58 and AIEC strain LF82 the two most important bacterial species involved in the pathogenesis of such diseases (Kukavica-Ibrulj et al. 2008; Darfeuille-Michaud et al. 2004). Moreover, since epithelial cells are considered as non-professional phagocytes, we evaluated whether Lf could modulate the expression of ferroportin (Fpn) (Cairo et al. 2011; Recalcati et al. 2012) the only known cellular iron exporter from cells (De Domenico et al. 2007) whose expression is regulated by inflammation (Yang et al. 2002; Ludwiczek et al. 2003; Paesano et al. 2009; Paesano et al. 2010; Cairo et al. 2011) and pivotal in the control of intracellular bacteria multiplication and inflammation (Kasvosve 2013; Nairz et al. 2013).

Materials and methods Bacterial strains Pseudomonas aeruginosa LES58, highly virulent epidemic strain isolated from lung sputum of CF patient (Cheng et al. 1996), and Escherichia coli LF82, isolated from a chronic ileal lesion of a patient with CD, provided by Dr. Arlette Darfeuille-Michaud,

845

Universite´ d’Auvergne, France, prototype of adhesive/ invasive E. coli (AIEC) strains, were used. To check purity, the strains were streaked on Tryptose Soy agar (TSA) plates (Oxoid LTD, England) before the experiments. To prepare inoculum, P. aeruginosa LESB58 was grown overnight at 37 °C in Chemically defined medium (CDM) containing 100 lM ferric ions as (Fe)2(SO4)3 (IL100-CDM) (Berlutti et al. 2005). Broth cultures were diluted in CDM containing 1 lM ferric ions as (Fe)2(SO4)3 (Berlutti et al. 2005) to obtain 8.0 ± 0.5 9 107 colony forming units (CFUs)/ml. AIEC strain LF82 was grown in Brain Heart Infusion Broth (BHI, Oxoid, Rome, Italy) overnight at 37 °C. Epithelial models Primary human airway epithelium reconstituted in vitro with cells isolated from CF patients and cultivated on microporous filters (Transwells) of 6.5 mm of diameter, with a pore size of 0.4 lm at an air–liquid interface (MucilAir, Epithelix Sa`rl, Geneva, Switzerland) was used. Each epithelium contained about 4 9 105 cells. The epithelium model was composed of fully differentiated basal, goblet, and ciliated cells covered by mucus and it reproduced certain phenotypic characteristics of the CF epithelium as the production of thick and dense mucus. MucilAirTM Culture Medium (Epithelix Sa`rl) was used to maintain the epithelium during the experiments. Bronchial epithelial CFBE cell line, homozygous for the DF508-CFTR mutation (Cozens et al. 1994) was grown in Eagle’s minimum essential medium (MEM, Euroclone, Milan, Italy) supplemented with 10 % fetal bovine serum (FBS), 2 mM glutamine, 100 U/ml penicillin, and 100 lg/ml streptomycin (Euroclone, Milan, Italy) and maintained in a humidified atmosphere of 95 % air and 5 % CO2 at 37 °C. Human epithelial colorectal adenocarcinoma cellline (Caco-2) was grown in Dulbecco’s modified Eagle’s medium (DMEM, Euroclone, Milan, Italy), supplemented with 1 % penicillin/ streptomycin and 10 % fetal calf serum (FCS) and maintained in 5 % CO2 atmosphere with 95 % humidity at 37 °C. Caco-2 cells were seeded at a density of 1 9 104 cells/well in 24-well plates and allowed to differentiate for 15 days before infection. When required the differentiated Caco-2 monolayers were pre-stimulated for 48 h with 50 ng/ml of IFN-c.

123

846

Lactoferrin Highly purified bovine milk derivative Lf (bLf) was kindly provided by Morinaga Milk Industries Co., Ltd. (Tokyo, Japan). The purity of bLf was checked according to Siciliano et al. (1999). Lipopolysaccharide (LPS) contamination of bLf, estimated by Limulus Amebocyte assay (LAL Pyrochrome kit, PBI International, Italy), was equal to 0.5 ± 0.06 ng/mg of bLf. The bLf iron saturation was about 22 %. Before biological assays, bLf was sterilized by filtration (Millipore). Cytotoxic and antibacterial activity of bovine lactoferrin To evaluate bLf toxicity on CF epithelium, a total of 50 ll of bLf solutions containing from 0.1 to 1 mg/ml was added to the apical side of CF epithelium. After 4 h of incubation, the excess of the solutions was removed and the viability of the epithelium was assayed using resazurin (R7017, Sigma Aldrich, Italy) assay as indicated by the CF epithelium supplier. To evaluate bLf toxicity on differentiated Caco-2 monolayers, bLf solutions containing from 0.1 to 1 mg/ml were added to Caco-2 cells and after 4 and 24 h of incubation cell viability was determined by light microscopy after staining with 0.02 % Trypan blu (Sigma, Italy) and by cell counting. To evaluate the antibacterial activity of bLf, 5.0 ± 0.5 9 106 and of 1.0 ± 0.4 9 106 CFUs of both P. aeruginosa LESB58 and E. coli LF82 were treated with serial bLf dilutions (from 1.0 to 0. 05 mg/ml, final concentrations) for 4 and 24 h. After incubation at 37 °C, the number of CFUs was determined. Invasion assay CF bronchial epithelium was infected at the apical side with 50 ll of the inoculum containing about 5.0 ± 0.5 9 106 P. aeruginosa LESB58 bacteria (MOI 1:10) with and without 0.1 mg/ml bLf. After 4 h at 37 °C in CO2 atmosphere, the liquid excess was removed from the apical side of the epithelium to restore the air/liquid interface. The medium contained in the lower part of the transwells was recovered and fresh medium added. The infected epithelium was incubated for 20 h at 37 °C in CO2 atmosphere. After incubation, the epithelium was washed and lysed with

123

Biometals (2014) 27:843–856

50 ll of 1 % deoxycholic acid for 5 min at 37 °C. Cell lysates were diluted in sterile saline and plated on TSA agar to count adherent bacteria. Adhesion efficiency values were calculated as the percentage of the ratio between the numbers of adherent versus the inoculum. To determine the number of intracellular bacteria, after 24 h of incubation, the apical side of the epithelium was washed with sterile saline and a solution of amikacine (1 mg/ml) and ceftazydime (1 mg/ml) was added to kill the extracellular bacteria. Preliminary experiments showed that this antibiotic mixture killed 1.0 ± 0.5 9 107 UFCs of P. aeruginosa LESB58 in 1 h. After 1 h of incubation, the epithelium was washed and lysed. Cell lysates were diluted in sterile saline and plated on TSA agar to count live intracellular bacteria. Invasion efficiency values were calculated as the percentage of the ratio between the numbers of intracellular versus both infecting and adhering bacteria. The viability of the epithelium was assayed using resazurin assay as above specified. Uninfected and infected CFBE without or with bLf were used to estimate Fpn protein. CFBE cells were seeded in 6-well plates at a density of 5 9 105 cells/ well, in 2 ml of MEM supplemented with 100 lM penicillin-streptomycin, 2 mM glutamine and 10 % FBS. The plates were incubated for 48 h at 37 °C in an atmosphere of 95 % air and 5 % CO2. After incubation, the cells were washed twice with 5 ml of PBS without calcium and magnesium (Sigma Aldrich), and further incubated for 2 h in 5 ml of MEM without penicillin-streptomycin and FBS. In infection experiments, CFBE cells were infected with P. aeruginosa LESB 58 at MOI 1:10 with and without 0.1 mg/ml bLf. After 3 h of infection at 37 °C, supernatants were removed and CFBE cells were incubated with 1 mg/ml amikacine and 1 mg/ml ceftazidime for 1 h to kill the extracellular bacteria. After 1 h of incubation, CFBE cells were washed in PBS and fresh medium containing 0.5 mg/ml amikacine, 0.5 mg/ml ceftazidime, and 0.1 mg/ml bLf was added. After incubation for further 20 h, the cells were treated as detailed below (see ‘‘Western blot’’ section). Companion experiments were performed using uninfected CFBE cells. To evaluate invasion, after antibiotic treatment to kill extracellular bacteria, the cells were lysed, cell lysates were diluted in sterile saline and plated on TSA agar to count live intracellular bacteria. Caco-2 differentiated monolayers unstimulated or stimulated with 50 ng/ml IFN-c (Sigma, Italy) for

Biometals (2014) 27:843–856

847

48 h at 37 °C before the experiments, were infected with 1.0 ± 0.2 9 106 AIEC LF82 bacteria/ml in culture medium (MOI 1:10) with and without 0.1 mg/ml bLf. After 3 h of infection at 37 °C, supernatants were removed and Caco-2 cells were incubated with 100 lg/ml gentamicin or with 100 lg/ml gentamicin plus 0.1 mg/ml bLf for 1 h to kill the extracellular bacteria. After the final wash in PBS, Caco-2 differentiated monolayers were lysed by adding 0.5 ml of deionized water containing 0.1 % (vol/vol) Triton X-100. Cell lysates were diluted in sterile saline and plated on TSA agar to count live intracellular bacteria. E. coli LF82 strain was considered invasive when the mean invasion level was superior or equal to 0.1 % of the original inoculum. To evaluate intracellular bacterial survival, infected Caco-2 monolayers were incubated for further 20 h with 50 lg/ml gentamicin without or with 0.1 mg/ml bLf. Survival efficiency was quantified as change (fold) in CFUs at 24 h relative to 4 h. The viability of Caco-2 cells was determined by light microscopy after staining with 0.02 % Trypan blu (Sigma, Italy) and by cell counting at 4 h and at 24 h post-infection.

(1:10,000) and monoclonal anti-actin (BD) (1:10,000). After incubation with the appropriate secondary HRP-conjugated antibody, blots were developed with ECL Prime (GE Healthcare). Ferroportin levels were normalized on actin by densitometry analysis, performed with ImageJ.

Cytokine production

Preliminary experiments were carried out to establish the non-cytotoxic and non-bactericidal concentration of bLf. We found that a 0.1 mg/ml concentration did not affect the bacterial viability nor it exerted a cytotoxic activity, therefore this concentration was used. The P. aeruginosa ability to adhere to and invade CF bronchial epithelium through the apical side was assessed. P. aeruginosa was able to adhere to the apical side of CF bronchial epithelium as 3.3 ± 1.4 9 105 CFUs were found at 24 h. When bLf was added, a reduction, of adherent bacteria (2.3 ± 1.4 9 105 vs 3.3 ± 1.4 9 105 CFUs) was noticed although it was not significant (Fig. 1) (P [ 0.05). P. aeruginosa invaded the CF bronchial epithelium and the addition of bLf at the moment of the infection reduced the P. aeruginosa invasion efficiency of about 50 % (P \ 0.05) (Fig. 1). No significant differences of the epithelium viability after 4 h and at 24 h of incubation in all experimental conditions were observed. Concerning the invasive ability of E. coli strain LF82 at 4 h, different efficiencies were noticed in nonstimulated versus IFN-c- stimulated cells (Fig. 2). When the cells were stimulated with IFN-c, a

ELISA were performed to determine cytokine concentrations on culture medium of uninfected and infected epithelial models in the absence or presence of 0.1 mg/ml bLf by using Human IL-8, IL-6, and IL1b ELISA Max Deluxe Sets (BioLegend, San Diego, CA, USA). Western blot A total of 1.0 ± 0.5 9 107 cells was scraped in 1 mL of PBS containing PMSF 1 mM, harvested by centrifugation at 2,5009g for 5 min and stored at -80 °C. Cells were lysed in 300 ll MOPS 25 mM pH 7.4/NaCl 150 mM/Triton 1 % containing PMSF 1 mM, Leupeptin and Pepstatin 2 lM in ice for 1 h. Total protein content of samples was measured by the Bradford assay. For SDS-PAGE 20 lg of total protein were loaded per lane, for Fpn analysis SDS sample buffer containing DTT was added and samples were loaded without heat treatment. For Western blot analysis, primary antibodies used were: monoclonal anti-Fpn 31A5 (Amgen, described in Ross et al. 2012)

Statistical analysis Results are expressed as means ± standard deviations derived from at least three independent experiments. Student t test was used for Fnp expression and to compare adhesion and invasion efficiency between controls and infecting strains. A bilateral Wilcoxon signed rank test was utilized to compare adhesion and invasion efficiency of LF82 in presence and absence of bLf. In all cases, a P value less than or equal to 0.05 was considered statistically significant.

Results Effect of bLf on infection of epithelial models

123

848

Biometals (2014) 27:843–856

Fig. 1 Adhesion and invasion efficiencies of Pseudomonas aeruginosa LESB58 in primary human CF bronchial epithelium at 24 h. Bovine lactoferrin (bLf) (0.1 mg/ml) was added at the moment of infection and maintained during the first 4 h of incubation. a Adhesion percentage values (%) were calculated

as the ratio between the numbers of adherent versus the inoculum; b invasion percentage values (%) were calculated as the ratio between the numbers of intracellular versus both inoculum and adhering bacteria

significant increase in the mean invasion percentage was observed: 0.9 ± 0.47 % versus 0.15 ± 0.10 % (P = 0.002). Addition of bLf during the infection period significantly decreased fivefolds the number of invasive bacteria (mean invasion percentage of 0.03 ± 0.02 % vs 0.15 ± 0.10 %; P = 0.0002), whilst it was not able to significantly inhibit the LF82 invasion efficiency in IFN-c-stimulated cells (0.7 ± 0.37 % vs 0.9 ± 0.47 %). As shown in Fig. 2b, at 24 h LF82 was able to survive inside Caco-2 cells with a mean survival percentage of 76.17 ± 30.90 %. Both IFN-c and bLf treatments significantly reduced the LF82 intracellular survival efficiency: 0.79 ± 0.30 % in IFN-c stimulated cells and 17.68 ± 13.99 % in bLf treated cells versus control 76.17 ± 30.90 %. Considering the low intracellular survival efficiency of bacteria in IFN-c stimulated cells, the effect of bLf was not significant (2.40 ± 1.99 %). Cell viability did not change significantly at 4 h and at 24 h in all experimental conditions.

Effect of bLf on inflammatory response of uninfected and infected epithelial models

123

To evaluate the effect of bLf on inflammatory response of epithelial models, the levels of cytokines secreted in the supernatants were assessed by ELISA at 4 and 24 h (Tables 1, 2). Concerning the CF bronchial epithelium (Table 1), IL-8 was produced at high level even in the absence of bacterial infection. After 4 h of P. aeruginosa LESB58 infection an increased production, as compared to the baseline level was observed with a fivefold increase for IL-8 and a tenfold increase for IL6. At 4 h, the addition of bLf did not significantly influence the cytokine expression of uninfected as well as of infected epithelium. When the cytokine levels were examined at 24 h, significant increases for all cytokines were noticed in infected compared to uninfected CF epithelia (IL-6 and IL-1b more than 100 fold; IL-8 about threefold). Addition of bLf did

Biometals (2014) 27:843–856

849

Fig. 2 Invasion and survival efficiencies of Escherichia coli LF82 strain in differentiated Caco-2 monolayers. Caco-2 cells were stimulated with IFN-c (50 ng/ml) 48 h before the experiments; bovine lactoferrin (bLf) (0.1 mg/ml) was added at the moment of infection and maintained during the 24 h of

incubation. a Invasion percentage values were calculated as the ratio between the numbers of intracellular bacteria versus inoculum; Panel b: survival efficiency was quantified as change (fold) in CFUs at 24 h relative to 4 h

not influence the cytokine levels of uninfected epithelia, while significantly reduced IL-8, IL-6, and IL-1b in infected epithelia. Differentiated Caco-2 monolayers did not produce IL-6 at detectable level under all experimental conditions. Concerning IL-1b, this cytokine was expressed mostly by IFN-c-stimulated cells and the addition of bLf did not significantly affect the cytokine production (Table 2). Regarding IL-8, the addition of bLf to uninfected monolayers increased the cytokine level only in IFN-c-stimulated cells at 24 h (Table 2). The infection with LF82 induced a higher IL-8 production with respect to controls in all experimental conditions. Addition of bLf to infected cells induced a IL-8 significant increase only in IFN-c-stimulated cells at 4 h.

Effect of bLf on Fpn protein of uninfected and infected epithelial models To evaluate the effect of bLf on Fpn, the Fpn protein on uninfected and infected epithelial models was assessed by Western blot experiments. Concerning CF, CFBE cell line was used since the number of cells in each CF bronchial epithelium transwell was too low. Preliminary experiments showed that the invasion ability at 4 h of P. aeruginosa LESB58 in CFBE with and without bLf was similar to that observed in CF epithelium model. In particular, bLf reduced the invasion efficiency of about 50 % (data not shown). Fpn level in uninfected cells was not affected by the addition of bLf (Fig. 3). P. aeruginosa infection induced a significant increase

123

850

Biometals (2014) 27:843–856

Table 1 Cytokine production by human primary CF bronchial epithelium Treatment

Cytokines (pg/ml) IL-6 (pg/ml) 4h

24 h

IL-1 b (pg/ml)

IL-8 (pg/ml)

4h

4h

24 h

24 h

None (CTRL)

13.77 ± 3.2

218.7 ± 96.8

0.81 ± 0.1

1.2 ± 0.1

493.03 ± 61.2

1,520.4 ± 646.61

bLf (0.1 mg/ml)

16.98 ± 2.8

218.6 ± 213.1

5.01 ± 3.6

0.6 ± 0.05

567.18 ± 44.6

1,852.7 ± 4.0

P. aeruginosa LESB58

179.19 ± 45.2

30,010.6 ± 18,812.1

2.88 ± 1.0

141.3 ± 8.6

2,632.22 ± 320.7

4,550.7 ± 417.2

P. aeruginosa LESB58 plus bLf

206.55 ± 53.1

9,405.6 ± 5,900.2

3.28 ± 0.9

59.9 ± 18.4

2,979.42 ± 169.8

4,318.2 ± 149.9

P values bLf versus CTRL Infection versus CTRL

NS 0.031

NS 0.0237

NS NS

NS 0.0105

NS 0.025

NS 0.0365

Infection with bLf versus without bLf

NS

0.0418

NS

0.0104

NS

0.0265

Cytokine concentrations were assayed in the medium after 4 and 24 h of treatment. Bovine lactoferrin (bLf) (0.1 mg/ml) was added at the time of experiments; for details on infection protocol see ‘‘Materials and methods’’ section NS not significant

of Fpn with respect to control and the addition of bLf further increased the Fpn synthesis. The protein level of Fpn in unstimulated monolayers was the same under all experimental conditions (Fig. 4). In IFN-c stimulated monolayers, the LF82 infection induced a significant reduction of Fpn that was partially, although significantly, restored by the addition of bLf (Fig. 4).

Discussion We have evaluated the role of bLf on human primary CF bronchial epithelium and colonic epithelial cells infected with bacterial species involved in the pathogenesis of CF airway infection and CD, human pathologies characterized by destructive (pathological) inflammation. Firstly, the ability of bLf to affect bacterial entry in epithelial models was assessed. Concerning the CF bronchial epithelium model, P. aeruginosa LESB58 was not able to enter inside the CF epithelium cells in 3 h at variance with what occurs using epithelial cell lines (Berlutti et al. 2008; Valenti et al. 2011). This observation underlines the pivotal importance in the selection of the cellular model that should be as close as possible to the in vivo situation. Therefore, in the reported experiments the infection time was prolonged

123

to allow P. aeruginosa to invade the CF epithelium. P. aeruginosa showed both a good adhesion and invasion efficiencies (Fig. 1). In agreement with previous observations (Berlutti et al. 2008), bLf did not affect P. aeruginosa LESB58 adhesion, but significantly reduced the number of intracellular bacteria (Fig. 1). Concerning the intestinal epithelial model, to mimic inflammatory conditions of CD, characterized by a preponderance of Th1 associated cytokines (Strober and Fuss 2011), cell monolayers were stimulated with IFN-c. Different invasion efficiencies were recorded depending on IFN-c stimulation. In fact, IFN-c stimulated monolayers were invaded at significantly higher efficiency with respect to the unstimulated cells (Fig. 2). This result is not surprising, as it has been demonstrated that the ability of LF82 to adhere/invade to Caco-2 cells was significantly increased when the expression of CEACAM6 was induced after IFN-c stimulation (Barnich et al. 2007). The addition of bLf at the moment of infection significantly reduced the AIEC invasion ability in not IFN-c stimulated monolayers, whereas it was ineffective in reducing invasion in IFN-c-stimulated cells. When we examined the survival efficiency after 24 h of infection (Fig. 2), the addition of bLf significantly reduced the number of intracellular bacteria only in unstimulated cells. The IFN-c stimulation induced a more efficient killing activity and the number of

b

NS

NS

Infection with bLf versus without bLf

NS

NS

NS

1.7 ± 0.9 1.45 ± 1.0

1.2 ± 1.0

14 ± 3.8

NS

NS

NS

18.1 ± 9.4 10.7 ± 7.3

1.4 ± 0.8

13.7 ± 8.5

NS

NS

NS

15.7 ± 7.7 35.6 ± 25.6

25.4 ± 12.5

28.0 ± 18.0

b

a

NS

0.0159

0.0,154

1,011.7 ± 490.3 1,333.2 ± 503.1

25.0 ± 1.8

30.9 ± 29.9

4h

Unstimulated

IL-8 (pg/ml)

NS

0.0217

NS

2,876.4 ± 1,376.8 3,748.4 ± 1,045.6

2,783.0 ± 705.2

1,638.5 ± 1,261.2

24 h

0.045865

0.015864

NS

1,675.6 ± 433.7 2,105.8 ± 855.6

43.9 ± 14.6

79.4 ± 27.7

4h

IFN-c stimulated

Bovine lactoferrin (bLf) (0.1 mg/ml) bLf was added at the time of experiments; for details on infection protocol see ‘‘Materials and methods’’ section

Differentiated Caco-2 monolayers were pretreated for 48 h with IFN-c; for details on infection protocol see ‘‘Materials and methods’’ section

NS not significant

Cytokine concentrations were assayed in the medium after 4 and 24 h of treatment

NS

Infection versus CTRL

P values

9.7 ± 8.8 0.9 ± 1.1

1.4 ± 0.9

0.5 ± 0.7

24 h

4h

4h

24 h

IFN-c stimulateda

Unstimulated

IL-1b (pg/ml)

bLf versus CTRL

AIEC LF82 AIEC LF82 plus bLf (0.1 mg/ml)

bLf (0.1 mg/ml)

None (CTRL)

Treatment

Table 2 Cytokine production by differentiated Caco-2 monolayers

NS

0.0155

0.0161

4,801.6 ± 629.8 5,008.7 ± 641.3

4,595.9 ± 500.4

2,548.3 ± 8.3

24 h

Biometals (2014) 27:843–856 851

123

852

Biometals (2014) 27:843–856

Fig. 3 Western blot and densitometric evaluations of Fpn expression in CFBE bronchial cells at 24 h. Bovine lactoferrin (bLf) (0.1 mg/ml) was added at the moment of infection and maintained during the first 4 h of incubation. Left panel

representative western blot of Fpn and b actin; right panel mean values of densitometric evaluation of Fpn expression normalized on actin

intracellular bacteria was so low that a putative action of bLf was not highlighted. To the best of our knowledge, the reported data showed for the first time the bLf ability to interfere with the intracellular survival of bacterial pathogens. Since it has been shown that Lf is able to modulate the inflammatory response of epithelial cell lines (Berlutti et al. 2006; Valenti et al. 2011), here we evaluated the putative anti-inflammatory activity of bLf in uninfected and infected epithelial models. BLf differently modulated the inflammatory response of the two epithelial models. In particular, in CF bronchial epithelium model, bLf was unable to modulate the inflammatory response of uninfected and infected cells at 4 h (Table 1). At 24 h, the levels of pro-inflammatory cytokines expressed by infected CF epithelium model greatly increased and the addition of bLf significantly reduced the levels of all these cytokines (Table 1). These data confirm the potent anti-inflammatory function of bLf in CF airway models (Valenti et al. 2011).

Regarding the intestinal epithelial model, the only significant inflammatory response was related to IL-8 production (Table 2). In uninfected cells, the bLf addition did not significantly change the IL-8 level at 4 h, whereas surprisingly it significantly increased IL-8 in IFN-c stimulated cells at 24 h. LF82 infection induced significantly increased levels of IL-8 already in the early phase of infection in both IFN-c and in non-stimulated cells, even if at different extent (Table 2) (Sasaki et al. 2007; Huebner et al. 2011). Moreover, in IFN-c stimulated cells, the bLf addition was able to produce a further significant increase of the cytokine production (P = 0.016). This result suggest that in vivo the bLf-induced increase of IL-8 could contribute to the eradication of the acute infection by recruiting neutrophils in infection sites (Alzoghaibi 2005). Overall IFN-c stimulated monolayers were more responsive to bLf and to intracellular bacteria than the non-stimulated cells in the production of IL-8. These results suggest a putative synergic action of

123

Biometals (2014) 27:843–856

853

Fig. 4 Western blot and densitometric evaluations of Fpn expression in differentiated Caco-2 monolayers at 24 h. Caco-2 cells were stimulated with IFN-c (50 ng/ml) 48 h before the experiments; bovine lactoferrin (bLf) (0.1 mg/ml) was added at

the moment of infection and maintained during the 24 h of incubation. Left panel representative western blot of Fpn and b actin; right panel mean values of densitometric evaluation of Fpn expression normalized on actin

IFN-c and bLf in inducing a strong inflammatory response (Welsh et al. 2011). This is not surprising because IFN-c is a specific hallmark of CD in which high levels of lactoferrin in feces has been related to acute phase of the disease (Lewis 2011). In any case, the role of bLf in the inflammatory diseases in which an altered response of innate an acquired immunity play a pivotal role in the pathogenesis (Strober and Fuss 2011; Cohen and Prince 2012) should be further investigated. Taken together, the bLf abilities in reducing the intracellular survival of bacterial pathogens and in differently modulating the inflammatory response of epithelial models, lead us to hypothesize that bLf acts in different way depending on the epithelial model and infecting pathogen. To clarify the mechanism, we explored whether bLf could modulate the Fpn protein whose expression is regulated by inflammation and whose action, by diminishing the intracellular iron level, determines a non permissive environment for

intracellular pathogens (Yang et al. 2002; Ludwiczek et al. 2003; Paesano et al. 2009; Paesano et al. 2010; Cairo et al. 2011; Kasvosve 2013; Nairz et al. 2013). In CF epithelial model P. aeruginosa infection induced the up-regulation of Fpn protein, while in IFN-c stimulated monolayers AIEC infection resulted in decreased expression of Fpn (Figs. 3, 4). When the effect of bLf on Fpn protein was examined, results showed that bLf was ineffective in uninfected cells, while an up-expression of Fpn was observed in both CF and IFN-c stimulated cells (Figs. 3, 4). This is the first time that the bLf influence on Fpn protein in epithelial cells is shown. Although further experiments are needed, the reported data suggest that mechanism underlying the bLf modulating activity on inflammatory response in epithelial cells could be more complex than hypothesized so far (Valenti et al. 2011; Kim et al. 2012) and that a role of bLf in modulating cellular iron homeostasis should be taken into account.

123

854 Acknowledgments This work was granted by Italian Cystic Fibrosis Research Foundation (research project FFC#13/2013 adopted by Delegazione FFC di Palermo e di Vittoria Ragusa Catania 2) to FB and by Sapienza University of Rome to PV; thanks are due to Luigi Rosa for his enthusiastic technical assistance.

References Alzoghaibi MA (2005) Neutrophil expression and infiltration into Crohn’s intestine. Saudi J Gastroenterol 11:63–72 Barnich N, Carvalho FA, Glasser AL, Darcha C, Jantscheff P, Allez M, Peeters H, Bommelaer G, Desreumaux P, Colombel JF, Darfeuille-Michaud A (2007) CEACAM6 acts as a receptor for adherent-invasive E. coli, supporting ileal mucosa colonization in Crohn disease. J Clin Investig 117:1566–1574. doi:10.1172/JCI30504 Berlutti F, Morea C, Battistoni A, Sarli S, Cipriani P, Superti F, Ammendolia MG, Valenti P (2005) Iron availability influences aggregation, biofilm, adhesion and invasion of Pseudomonas aeruginosa and Burkholderia cenocepacia. Int J Immunopathol Pharmacol 18:661–670 Berlutti F, Schippa S, Morea C, Sarli S, Perfetto B, Donnarumma G, Valenti P (2006) Lactoferrin downregulates proinflammatory cytokines upexpressed in intestinal epithelial cells infected with invasive or non invasive Escherichia coli strains. Biochem Cell Biol 84:351–357 Berlutti F, Superti F, Nicoletti M, Morea C, Frioni A, Ammendolia MG, Battistoni A, Valenti P (2008) Bovine lactoferrin inhibits the efficiency of invasion of respiratory A549 cells of different iron-regulated morphological forms of Pseudomonas aeruginosa and Burkholderia cenocepacia. Int J Immunopathol Pharmacol 21:51–59 Boucher RC (2002) An overview of the pathogenesis of cystic fibrosis lung disease. Adv Drug Deliv Rev 54:1359–1371. doi:10.1016/S0169-409X(02)00144-8 Cairo G, Recalcati S, Mantovani A, Locati M (2011) Iron trafficking and metabolism in macrophages: contribution to the polarized phenotype. Trends Immunol 32:241–247. doi:10.1016/j.it.2011.03.007 Cario E, Rosenberg IM, Brandwein SL, Beck PL, Reinecker HC, Podolsky DK (2000) Lipopolysaccharide activates distinct signaling pathways in intestinal epithelial cell lines expressing Toll-like receptors. J Immunol 164:966–972 Chassaing B, Koren O, Carvalho FA, Ley RE, Gewirtz AT (2013) AIEC pathobiont instigates chronic colitis in susceptible hosts by altering microbiota composition. Gut. doi:10.1136/gutjnl-2013-304909 Cheng K, Smyth RL, Govan JR, Doherty C, Winstanley C, Denning N, Heaf DP, van Saene H, Hart CA (1996) Spread of beta-lactam-resistant Pseudomonas aeruginosa in a cystic fibrosis clinic. Lancet 348:639–642. doi:10.1016/ S0140-6736(96)05169-0 Cohen TS, Prince A (2012) Cystic fibrosis: a mucosal immunodeficiency syndrome. Nat Med 18:509–519. doi:10. 1038/nm.2715 Cozens AL, Yezzi MJ, Kunzelmann K, Ohrui T, Chin L, Eng K, Finkbeiner WE, Widdicombe JH, Gruenert DC (1994) CFTR expression and chloride secretion in polarized

123

Biometals (2014) 27:843–856 immortal human bronchial epithelial cells. Am J Respir Cell Mol Biol 10:38–47. doi:10.1165/ajrcmb.10.1.7507342 Curran C, Demick KP, Mansfield JM (2006) Lactoferrin activates macrophages via TLR4-dependent and -independent signaling pathways. Cell Immunol 242:23–30. doi:10. 1016/j.cellimm.2006.08.006 Darfeuille-Michaud A, Boudeau J, Bulois P, Neut C, Glasser AL, Barnich N, Bringer MA, Swidsinski A, Beaugerie L, Colombel JF (2004) High prevalence of adherent-invasive Escherichia coli associated with ileal mucosa in Crohn’s disease. Gastroenterology 127:412–421. doi:10.1053/j. gastro.2004.04.061 De Domenico I, Ward DM, Musci G, Kaplan J (2007) Evidence for the multimeric structure of ferroportin. Blood 109:2205–2209. doi:10.1182/blood-2006-06-032516 Ghio AJ, Piantadosi CA, Wang X, Dailey LA, Stonehuerner JD, Madden MC, Yang F, Dolan KG, Garrick MD, Garrick LM (2005) Divalent metal transporter-1 decreases metal-related injury in the lung. Am J Physiol Lung Cell Mol Physiol 289:L460–L467. doi:10.1152/ajplung.00154.2005 Glasser AL, Boudeau J, Barnich N, Perruchot MH, Colombel JF, Darfeuille-Michaud A (2001) Adherent invasive Escherichia coli strains from patients with Crohn’s disease survive and replicate within macrophages without inducing host cell death. Infect Immunol 69:5529–5537. doi:10. 1128/IAI.69.9.5529-5537.2001 Hayworth JL, Kasper KJ, Leon-Ponte M, Herfst CA, Yue D, Brintnell WC, Mazzuca DM, Heinrichs DE, Cairns E, Madrenas J, Hoskin DW, McCormick JK, Haeryfar SM (2009) Attenuation of massive cytokine response to the staphylococcal enterotoxin B superantigen by the innate immunomodulatory protein lactoferrin. Clin Exp Immunol 157:60–70. doi:10.1111/j.1365-2249.2009.03963.x Huebner C, Ding Y, Petermann I, Knapp C, Ferguson LR (2011) The probiotic Escherichia coli Nissle 1917 reduces pathogen invasion and modulates cytokine expression in Caco2 cells infected with Crohn’s disease-associated E. coli LF82. Appl Environ Microbiol 77:2541–2544. doi:10. 1128/AEM.01601-10 Kasvosve I (2013) Effect of ferroportin polymorphism on iron homeostasis and infection. Clin Chim Acta 1(416):20–25. doi:10.1016/j.cca.2012.11.013 Khan TZ, Wagener JS, Bost T, Martinez J, Accurso FJ, Riches DW (1995) Early pulmonary inflammation in infants with cystic fibrosis. Am J Respir Crit Care Med 151:1075–1082. doi:10.1164/ajrccm.151.4.7697234 Kim CW, Lee TH, Park KH, Choi SY, Kim J (2012) Human lactoferrin suppresses TNF-a-induced intercellular adhesion molecule-1 expression via competition with NF-jB in endothelial cells. FEBS Lett 586:229–234. doi:10.1016/j. febslet.2011.12.011 Komatsu A, Satoh T, Wakabayashi H, Ikeda F (2013) Effects of bovine lactoferrin to oral Candida albicans and Candida glabrata isolates recovered from the saliva in elderly people. Odontology Kukavica-Ibrulj I, Bragonzi A, Paroni M, Winstanley C, Sanschagrin F, O’Toole GA, Levesque RC (2008) In vivo growth of Pseudomonas aeruginosa strains PAO1 and PA14 and the hypervirulent strain LESB58 in a rat model of chronic lung infection. J Bacteriol 190:2804–2813. doi:10.1128/JB.01572-07

Biometals (2014) 27:843–856 Latorre D, Berlutti F, Valenti P, Gessani S, Puddu P (2012) LF immunomodulatory strategies: mastering bacterial endotoxin. Biochem Cell Biol 90:269–278. doi:10.1139/o11059 Lee TW, Brownlee KG, Conway SP, Denton M, Littlewood JM (2003) Evaluation of a new definition for chronic Pseudomonas aeruginosa infection in cystic fibrosis patients. J Cyst Fibros 2:29–34. doi:10.1016/S1569-1993(02)00141-8 Legrand D (2012) Lactoferrin, a key molecule in immune and inflammatory processes. Biochem Cell Biol 90:252–268. doi:10.1139/o11-056 Lewis JD (2011) The utility of biomarkers in the diagnosis and therapy of inflammatory bowel disease. Gastroenterology 140(6):1817–1826 Ludwiczek S, Aigner E, Theurl I, Weiss G (2003) Cytokinemediated regulation of iron transport in human monocytic cells. Blood 101:4148–4154. doi:10.1182/blood-2002-082459 Martinez-Medina M, Aldeguer X, Lopez-Siles M, Gonza´lezHuix F, Lopez-Oliu C, Dahbi G, Blanco JE, Blanco J, Garcia-Gil LJ, Darfeuille-Michaud A (2009) Molecular diversity of Escherichia coli in the human gut: new ecological evidence supporting the role of adherent-invasive E. coli (AIEC) in Crohn’s disease. Inflamm Bowel Dis 15:872–882. doi:10.1002/ibd.20860 Moreau-Marquis S, Bomberger JM, Anderson GG, SwiateckaUrban A, Ye S, O’Toole GA, Stanton BA (2008) The DeltaF508-CFTR mutation results in increased biofilm formation by Pseudomonas aeruginosa by increasing iron availability. Am J Physiol Lung Cell Mol Physiol 295:L25–L37. doi:10.1152/ajplung.00391 Nairz M, Schleicher U, Schroll A, Sonnweber T, Theurl I, Ludwiczek S, Talasz H, Brandacher G, Moser PL, Muckenthaler MU, Fang FC, Bogdan C, Weiss G (2013) Nitric oxide-mediated regulation of ferroportin-1 controls macrophage iron homeostasis and immune function in Salmonella infection. J Exp Med 210(5):855–873 Nichols D, Chmiel J, Berger M (2007) Chronic inflammation in the cystic fibrosis lung: alterations in inter- and intracellular signalling. Clin Rev Allergy Immunol 34:146–162 O’Sullivan BP, Flume P (2009) The clinical approach to lung disease in patients with cystic fibrosis. Semin Respir Crit Care Med 30:505–513. doi:10.1055/s-0029-1238909 Paesano R, Pietropaoli M, Gessani S, Valenti P (2009) The influence of lactoferrin, orally administered, on systemic iron homeostasis in pregnant women suffering of iron deficiency and iron deficiency anemia. Biochimie 91:44–51. doi:10.1016/j.biochi.2008.06.004 Paesano R, Berlutti F, Pietropaoli M, Goolsbee W, Pacifici E, Valenti P (2010) Lactoferrin efficacy versus ferrous sulfate in curing iron disorders in pregnant and non pregnant women. Int J Immunopathol Pharmacol 23:577–587 Pfefferkorn MD, Boone JH, Nguyen JT, Juliar BE, Davis MA, Parker KK (2010) Utility of fecal lactoferrin in identifying Crohn disease activity in children. J Pediatr Gastroenterol Nutr 51:425–428. doi:10.1097/MPG.0b013e3181d67e8f Puddu P, Carollo MG, Belardelli F, Valenti P, Gessani S (2007) Role of endogenous interferon and LPS in the immunomodulatory effects of bovine lactoferrin in murine peritoneal macrophages. J Leukoc Biol 82:347–353. doi:10. 1189/jlb.1106688

855 Puddu P, Latorre D, Carollo M, Catizone A, Ricci G, Valenti P, Gessani S (2011) Bovine lactoferrin counteracts Toll-like receptor mediated activation signals in antigen presenting cells. PLoS ONE 6:e22504. doi:10.1371/journal.pone. 0022504 Ranganathan SC, Parsons F, Gangell C, Brennan S, Stick SM, Sly PD (2011) Evolution of pulmonary inflammation and nutritional status in infants and young children with cystic fibrosis. Thorax 66:408–413. doi:10.1136/thx.2010. 139493 Recalcati S, Locati M, Gammella E, Invernizzi P, Cairo G (2012) Iron levels in polarized macrophages: regulation of immunity and autoimmunity. Autoimmun Rev 11:839–883. doi:10.1016/j.autrev.2012.03.003 Reid DW, Lam QT, Schneider H, Walters EH (2004) Airway iron and iron-regulatory cytokines in cystic fibrosis. Eur Respir J 24:286–291. doi:10.1183/09031936.04.00104803 Reid DW, Carroll V, O’May C, Champion A, Kirov SM (2007) Increased airway iron as a potential factor in the persistence of Pseudomonas aeruginosa infection in cystic fibrosis. Eur Respir J 30:286–292. doi:10.1183/09031936. 00154006 Rolhion N, Darfeuille-Michaud A (2007) Adherent-invasive Escherichia coli in inflammatory bowel disease. Inflamm Bowel Dis 13:1277–1283 Ross SL, Tran L, Winters A, Lee KJ, Plewa C, Foltz I, King C, Miranda LP, Allen J, Beckman H, Cooke KS, Moody G, Sasu BJ, Nemeth E, Ganz T, Molineux G, Arvedson TL (2012) Molecular mechanism of hepcidin-mediated ferroportin internalization requires ferroportin lysines, not tyrosines or JAK-STAT. Cell Metab 15(6):905–917. doi:10. 1016/j.cmet.2012.03.017 Sagel SD, Sontag MK, Accurso FJ (2009) Relationship between antimicrobial proteins and airway inflammation and infection in cystic fibrosis. Pediatr Pulmonol 44:402–409. doi:10.1002/ppul.21028 Sasaki M, Sitaraman SV, Babbin BA, Gerner-Smidt P, Ribot EM, Garrett N, Alpern JA, Akyildiz A, Theiss AL, Nusrat A, Klapproth JM (2007) Invasive Escherichia coli are a feature of Crohn’s disease. Lab Invest 87:1042–1054. doi:10.1038/labinvest.3700661 Siciliano R, Rega B, Marchetti M, Seganti L, Antonini G, Valenti P (1999) Bovine lactoferrin peptidic fragments involved in inhibition of herpes simplex virus type 1 infection. Biochem Biophys Res Commun 264:19–23. doi:10.1006/bbrc.1999.1318 Strober W, Fuss IJ (2011) Pro-inflammatory cytokines in the pathogenesis of IBD. Gastroenterology 140:1756–1767. doi:10.1053/j.gastro.2011.02.016 Tirouvanziam R, Khazaal I, Peault B (2002) Primary inflammation in human cystic fibrosis small airways. Am J Physiol Lung Cell Mol Physiol 283:L445–L451. doi:10. 1152/ajplung.00419.2001 Tunney MM, Field TR, Moriarty TF, Patrick S, Doering G, Muhlebach MS, Wolfgang MC, Boucher R, Gilpin DF, McDowell A, Elborn JS (2008) Detection of anaerobic bacteria in high numbers in sputum from patients with cystic fibrosis. Am J Respir Crit Care Med 177:995–1001. doi:10.1164/rccm.200708-1151OC Valenti P, Catizone A, Pantanella F, Frioni A, Natalizi T, Tendini M, Berlutti F (2011) Lactoferrin decreases

123

856 inflammatory response by cystic fibrosis bronchial cells invaded with Burkholderia cenocepacia iron-modulated biofilm. Int J Immunopathol Pharmacol 24:1057–1068 Vora P, Youdim A, Thomas LS, Fukata M, Tesfay SY, Lukasek K, Michelsen KS, Wada A, Hirayama T, Arditi M, Abreu MT (2004) Beta-defensin-2 expression is regulated by TLR signaling in intestinal epithelial cells. J Immunol 173:5398–5405 Wang G (2010) State-dependent regulation of cystic fibrosis transmembrane conductance regulator (CFTR) gating by a high affinity Fe3? bridge between the regulatory domain and cytoplasmic loop 3. J Biol Chem 285:40438–40447. doi:10.1074/jbc.M110.161497

123

Biometals (2014) 27:843–856 Welsh KJ, Hwang SA, Boyd S, Kruzel ML, Hunter RL, Actor JK (2011) Influence of oral lactoferrin on Mycobacterium tuberculosis induced immunopathology. Tuberculosis 1:S105–S113. doi:10.1016/j.tube.2011.10.019 Wiesner J, Vilcinskas A (2010) Antimicrobial peptides: the ancient arm of the human immune system. Virulence 1:440–464. doi:10.4161/viru.1.5.12983 Yang F, Liu XB, Quinones M, Melby PC, Ghio A, Haile DJ (2002) Regulation of reticuloendothelial iron transporter MTP1 (Slc11a3) by inflammation. J Biol Chem 277:39786– 39791