murine antigen-induced inflammation—a model for studying induction, resolution and the adaptive...

10
Research paper Murine antigen-induced inammationA model for studying induction, resolution and the adaptive phase of inammation Valgerdur Tomasdottir a,b , Arnor Vikingsson d , Ingibjorg Hardardottir a,1 , Jona Freysdottir b,c,d, ,1 a Department of Biochemistry and Molecular Biology, Faculty of Medicine, Biomedical Center, University of Iceland, Reykjavik, Iceland b Department of Immunology, Faculty of Medicine, Biomedical Center, University of Iceland, Reykjavik, Iceland c Department of Immunology, Landspitali - The National University Hospital of Iceland, Reykjavik, Iceland d Center for Rheumatology Research, Landspitali - The National University Hospital of Iceland, Reykjavik, Iceland article info abstract Article history: Received 31 January 2014 Received in revised form 27 August 2014 Accepted 22 September 2014 Available online xxxx Murine zymosan-induced peritonitis is the model most frequently used to study resolution of inflammation. However, the antigen-induced peritonitis model may be better suited for studying resolution of inflammation and the adaptive phase that follows. The objective of this study was to provide an evaluation of the kinetics of cells and mediators during induction, resolution and the adaptive immune phases of a murine antigen-induced inflammation. Female C57BL/6 mice were immunized twice subcutaneously with mBSA and three weeks after the initial immunization they were injected intraperitoneally (i.p.) with mBSA, which induced peritonitis. Peritoneal cells were counted and expression of surface molecules and chemokine receptors analyzed with flow cytometry. Chemokine and cytokine concentrations in peritoneal fluid were determined by ELISA. Two neutrophil populations, differing in size and granularity and slightly in expression of surface molecules, were observed in the peritoneal cavity after induction of inflammation. Macrophages disappeared from the peritoneal cavity following i.p. administration of mBSA but appeared again as they differentiated from recruited monocytes and peaked in numbers at 48 h. At that time point, two distinct populations of macrophages were present in the peritoneal cavity; one with high expression of F4/80, also expressing the atypical chemokine receptor D6 as well as CCR7; the other expressing low levels of F4/80 and also expressing CD11c and CD138. Eosinophils appeared in the peritoneum 3 h following i.p. administration of mBSA and peaked at 48 h. At that time point they had upregulated their expression of CCR3 but decreased their expression of CD11b. Peritoneal levels of CCL11 peaked at 6 h and may have led to recruitment of the eosinophils. NK cells and T cells peaked at 48 h, whereas B cells peaked at 5 days, with the majority being B1 cells. Peritoneal concentrations of pro-inflammatory cytokines (IL-β and IL-6) and chemokines (CCL2 and CCL3) peaked at 3 h, whereas IL-1ra peaked at 6 h, sTNF-R at 24 h and sIL-6R and TGF-β at 48 h. The results show kinetic alterations in cell populations and mediators in a murine model that may be an excellent model to study initiation and resolution of inflammation and the following adaptive phase. © 2014 Elsevier B.V. All rights reserved. Keywords: Antigen-induced inflammation Murine model Induction of inflammation Resolution of inflammation 1. Introduction The inflammatory response consists of an initial induc- tion phase that evolves into a resolution phase. The fine tuning of these responses is essential for maximizing the Journal of Immunological Methods xxx (2014) xxxxxx Corresponding author at: Department of Immunology and Centre for Rheumatology Research, Landspitali - The National University Hospital of Iceland, IS-101 Reykjavik, Iceland. Tel.: +354 8612056; fax: +354 5434828. E-mail address: [email protected] (J. Freysdottir). 1 Joint last authorship. JIM-11913; No of Pages 10 http://dx.doi.org/10.1016/j.jim.2014.09.004 0022-1759/© 2014 Elsevier B.V. All rights reserved. Contents lists available at ScienceDirect Journal of Immunological Methods journal homepage: www.elsevier.com/locate/jim Please cite this article as: Tomasdottir, V., et al., Murine antigen-induced inammationA model for studying induction, resolution and the adaptive phase of inammation, J. Immunol. Methods (2014), http://dx.doi.org/10.1016/j.jim.2014.09.004

Upload: jona

Post on 09-Feb-2017

214 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Murine antigen-induced inflammation—A model for studying induction, resolution and the adaptive phase of inflammation

Journal of Immunological Methods xxx (2014) xxx–xxx

JIM-11913; No of Pages 10

Contents lists available at ScienceDirect

Journal of Immunological Methods

j ourna l homepage: www.e lsev ie r .com/ locate / j im

Research paper

Murine antigen-induced inflammation—A model for studyinginduction, resolution and the adaptive phase of inflammation

Valgerdur Tomasdottir a,b, Arnor Vikingsson d, Ingibjorg Hardardottir a,1, Jona Freysdottir b,c,d,⁎,1

a Department of Biochemistry and Molecular Biology, Faculty of Medicine, Biomedical Center, University of Iceland, Reykjavik, Icelandb Department of Immunology, Faculty of Medicine, Biomedical Center, University of Iceland, Reykjavik, Icelandc Department of Immunology, Landspitali - The National University Hospital of Iceland, Reykjavik, Icelandd Center for Rheumatology Research, Landspitali - The National University Hospital of Iceland, Reykjavik, Iceland

a r t i c l e i n f o

⁎ Corresponding author at: Department of ImmunRheumatology Research, Landspitali - The National UIceland, IS-101 Reykjavik, Iceland. Tel.: +354 8612056;

E-mail address: [email protected] (J. Freysdottir).1 Joint last authorship.

http://dx.doi.org/10.1016/j.jim.2014.09.0040022-1759/© 2014 Elsevier B.V. All rights reserved.

Please cite this article as: Tomasdottir, V., etand the adaptive phase of inflammation, J.

a b s t r a c t

Article history:Received 31 January 2014Received in revised form 27 August 2014Accepted 22 September 2014Available online xxxx

Murine zymosan-induced peritonitis is the model most frequently used to study resolution ofinflammation. However, the antigen-induced peritonitis model may be better suited for studyingresolution of inflammation and the adaptive phase that follows. The objective of this study was toprovide an evaluation of the kinetics of cells and mediators during induction, resolution and theadaptive immune phases of a murine antigen-induced inflammation.Female C57BL/6 mice were immunized twice subcutaneously with mBSA and three weeks afterthe initial immunization they were injected intraperitoneally (i.p.) with mBSA, which inducedperitonitis. Peritoneal cells were counted and expression of surface molecules and chemokinereceptors analyzed with flow cytometry. Chemokine and cytokine concentrations in peritonealfluid were determined by ELISA.Two neutrophil populations, differing in size and granularity and slightly in expression of surfacemolecules, were observed in the peritoneal cavity after induction of inflammation. Macrophagesdisappeared from the peritoneal cavity following i.p. administration ofmBSA but appeared again asthey differentiated from recruited monocytes and peaked in numbers at 48 h. At that time point,two distinct populations of macrophages were present in the peritoneal cavity; one with highexpression of F4/80, also expressing the atypical chemokine receptor D6 as well as CCR7; the otherexpressing low levels of F4/80 and also expressing CD11c and CD138. Eosinophils appeared in theperitoneum 3 h following i.p. administration of mBSA and peaked at 48 h. At that time point theyhad upregulated their expression of CCR3 but decreased their expression of CD11b. Peritoneallevels of CCL11 peaked at 6 h and may have led to recruitment of the eosinophils. NK cells and Tcells peaked at 48 h, whereas B cells peaked at 5 days, with the majority being B1 cells. Peritonealconcentrations of pro-inflammatory cytokines (IL-β and IL-6) and chemokines (CCL2 and CCL3)peaked at 3 h, whereas IL-1ra peaked at 6 h, sTNF-R at 24 h and sIL-6R and TGF-β at 48 h.The results show kinetic alterations in cell populations andmediators in amurinemodel thatmaybe an excellent model to study initiation and resolution of inflammation and the followingadaptive phase.

© 2014 Elsevier B.V. All rights reserved.

Keywords:Antigen-induced inflammationMurine modelInduction of inflammationResolution of inflammation

ology and Centre forniversity Hospital offax: +354 5434828.

al., Murine antigen-induImmunol. Methods (201

1. Introduction

The inflammatory response consists of an initial induc-tion phase that evolves into a resolution phase. The finetuning of these responses is essential for maximizing the

ced inflammation—Amodel for studying induction, resolution4), http://dx.doi.org/10.1016/j.jim.2014.09.004

Page 2: Murine antigen-induced inflammation—A model for studying induction, resolution and the adaptive phase of inflammation

2 V. Tomasdottir et al. / Journal of Immunological Methods xxx (2014) xxx–xxx

benefits and minimizing the harm ensuing from activationof the inflammatory cascade. Although the inflammatoryresponse is customarily initiated by activation of the innateimmune system, adaptive immunity can serve a valuable role indiminishing inflammatory responses and accelerate recoveryfollowing re-exposure to an antigen.

Induction and resolution of inflammation has beenthoroughly studied in the zymosan-induced peritonitismodel (reviewed in (Serhan, 2007)), as well as in allergicairway inflammation (Haworth et al., 2011; Rogerio et al.,2012). Zymosan triggers the inflammatory response bybinding to toll-like receptor (TLR)-2 on tissue monocytes/macrophages, thereby inducing cytokine and chemokinesecretion. Marked but transient influx of neutrophils intothe peritoneum is followed by infiltration and activationof monocytes/macrophages. Initially, the macrophagesare pro-inflammatory (M1 macrophages), secreting tumornecrosis factor (TNF)-α, interleukin (IL)-1 and IL-6 andvigorously phagocytosing foreign antigens and apoptoticneutrophils, but gradually their phenotype changes toan anti-inflammatory or pro-resolution type (M2 or rMmacrophages, respectively) (Gilroy et al., 2004; Schif-Zucket al., 2011), characterized by predominant production ofpro-resolution lipid mediators (i.e., lipoxins, resolvins,protectins and maresins) and anti-inflammatory cytokines(e.g. IL-10 and transforming growth factor (TGF)-ß)(reviewed in (Ariel and Timor, 2013)). This resolutionprocess may be further strengthened by the emergenceof a recently described resolution-promoting eosinophils(Yamada et al., 2011). Eventually, the inflammatory lesionrecedes and post inflammation homeostasis is restored.

Induction of inflammation within the sterile peritonealcavity represents an ideal location to study the evolution ofthe inflammatory response due to the easy access to theperitoneal exudate for analysis of cells and soluble media-tors. Therefore, zymosan-induced peritonitis is an excellentmodel to study an intense self-limited acute inflammationresulting from TLR activation. However, the zymosan-induced peritonitis model entails little or no activation ofthe adaptive immune response (Kolaczkowska et al., 2008)and may, therefore, not accurately reflect the kinetics of theinduction and resolution phases during naturally occurringinfections or during immune reactions that have thepotential to evolve into chronic inflammation, includingautoimmune processes. In the methylated BSA (mBSA)-induced peritonitis model, both the innate and the adaptiveelements of the immune response are initially activated byimmunizing subcutaneously (s.c.) with mBSA and completeFreund´s adjuvant. Peritonitis is subsequently induced byintraperitoneal (i.p.) injection of mBSA without an adju-vant, resulting in reactivation of the adaptive immuneresponse. In previous studies, Cook et al. described tempo-ral changes in the number of neutrophils and macrophagesin mBSA-induced peritonitis (Cook et al., 2003, 2004) butonly provided analysis of surface markers and solublemediators within the peritoneal cavity on day 4 followinginduction of inflammation. In the present study, we providea detailed analysis of cells and soluble mediators during theinduction and resolution phases as well as the subsequentadaptive phase of inflammation in murine mBSA-inducedperitonitis.

Please cite this article as: Tomasdottir, V., et al., Murine antigen-induand the adaptive phase of inflammation, J. Immunol. Methods (201

2. Materials and methods

2.1. Mice

All animal procedures were approved by the ExperimentalAnimal Committee, Ministry for the Environment in Iceland(#0507-1503) and compliedwith NRC´s Guide for the Care andUse of Laboratory Animals. Female C57BL/6mice, weighing 18–20 g, were obtained from Taconic Europe (Ejby, Denmark).They were housed 8 per cage with a 12 h light/dark cycle at23–25 °C and 45–55% humidity. Mice were acclimated for1 week prior to initiation of the experiments. Mice had freeaccess to food and water.

2.2. Induction of peritonitis

mBSA (Sigma Aldrich, St. Louis, MO) was dissolved in waterand 10× PBS added to give the final concentration of 1× PBS.Mice were immunized s.c. at the base of the tail with 100 μg ofmBSA emulsified in equal volume of CFA (Sigma Aldrich) untilwhite and thick. The total injection volume was 50 μl. Twoweeks later, themicewere given a booster injection of 100 μg ofmBSA in incomplete Freund‘s adjuvant (IFA) (Sigma Aldrich).Three weeks after the initial immunization, mice werechallenged i.p. with 100 μg of mBSA in 50 μl of saline. Beforeand at several time points after peritonitis induction (3 h, 6 h,12 h, 24 h, 48 h, 5 days and 10 days), mice were anesthetizedwith a mixture of hypnorm (VetaPharma Ltd, Leeds, UK),dormicum (Roche, Basel, Switzerland) and sterilewater (1:1:2)and killed by cervical dislocation.

2.3. Collection of serum, peritoneal lavage and spleen

Serum was collected and stored at −70 °C. Peritonealexudate was obtained by injecting 1.5 ml of cold phosphate-buffered saline (PBS), without calcium or magnesium, intothe peritoneal cavity and collecting the fluid. Cells andsupernatant were separated by centrifugation. The super-natant was collected and stored at −70 °C. Spleen wascollected and pushed through a sieve in order to obtainsingle cell suspension. Red blood cells were lysed using ACKlysing buffer (0.15 M NH4Cl, 1 mM KHCO3 and 0.1 mMNa2EDTA). Peritoneal and spleen cells were washed twicewith PBS and resuspended in FACS buffer (PBS containing 1%BSA, 0.2 mM EDTA and 0.1% NaN3) and counted by Countessautomated cell counter (Invitrogen, Paisley, UK).

2.4. Characterization of cells by flow cytometry

Peritoneal and spleen cells (0.3 × 106) were incubated with2% normal rat:normal mouse serum (1:1) (AbD Serotec,Kidlington, UK) for 20 min. Cells were stained withfluorochrome-labeled monoclonal antibodies (mabs) againstGr1, F4/80, CD115, CD90.2, NK1.1, CD4, CD8, B220, IgD, CD5(eBioscience, San Diego, CA), Ly6G (clone 1A8), CD11b, F4/80(BD Bioscience, San Jose, CA), CCR3, CXCR2, D6, CCR7 (R&DSystems, Abington, UK) and CD11c (MBL-Nordic Biosite, Taby,Sweden). Appropriate isotype controls were used to set thequadrants and evaluate background staining. Cells werewashed twice, resuspended in FACS buffer, and 10,000 cellswere collected on FACScalibur (BD Biosciences). Data were

ced inflammation—Amodel for studying induction, resolution4), http://dx.doi.org/10.1016/j.jim.2014.09.004

Page 3: Murine antigen-induced inflammation—A model for studying induction, resolution and the adaptive phase of inflammation

3V. Tomasdottir et al. / Journal of Immunological Methods xxx (2014) xxx–xxx

analyzed using FlowJo software (Tree Star Inc, Ashland, OR).The macrophages were identified as cells expressing F4/80 andCD11b; the monocytes as cells expressing CD115 and notF4/80; the eosinophils as cells expressing CCR3; thegranulocytes as cells expressing Ly6G, CD11b and CXCR2; andthe lymphocytes as cells expressing B220, CD90.2 and NK1.1.

2.5. Measurement of BSA-specific IgM and IgG antibodies in serum

BSA-specific IgM and IgG antibodies were measured inserum using indirect ELISA. Maxisorp plates (Nunc, Invitrogen)were coated overnight at 4 °C with 50 μg/ml BSA (Sigma-Aldrich) in bicarbonate buffer (pH 9.6). Standard (pooledserum from mBSA immunized mice) and sera, diluted from1/10,000 for measuring IgG anti-BSA antibodies and from 1/100 for IgM antibodies, were incubated for 2 h at roomtemperature and further incubated with horseradish peroxi-dase (HRP)-labeled goat anti-mouse IgM and IgG (SouthernBiotech, Birmingham, AL) for 2 h. The substrate 3,3′,5,5′-tetramethylbenzidine (Kirkegaard & Perry Laboratories,Gaithersburg, MD) was added and the color reaction stoppedwith 0.18 M H2SO4. The results are expressed as arbitrary unitscalculated from standard curves made from serial dilution ofthe standard.

2.6. Cytokine and chemokine analysis

Concentrations of cytokines (IL-1β, IL-6, IL-10, IL-12 andTGF-β), chemokines (CCL2, CCL3 and CCL11) and solublecytokine receptors (IL-1ra, sTNF-R and sIL-6R) were measuredin peritoneal fluid using Duoset ELISA kits (R&D Systems).

2.7. Statistical analysis

Results are expressed as mean values ± standard error ofthe mean (SEM). One-way ANOVA followed by Tukey's posthoc test was used to determine whether the effects of mBSAadministration were statistically significant. Where data werenot normally distributed, one-way ANOVA of ranks followedby Dunn's post hoc test was used to determine whether groupmedians differed, but means ± SEM shown for clarity.Differences between means or medians were consideredsignificant if P b 0.05. Statistical analysis was performed usingSigmaStat software, version 3.2 (Systat software Inc, Chicago,IL).

3. Results

3.1. Cell populations

Macrophages and lymphocytes were the main cell types inthe peritoneal cavity prior to i.p. injection of mBSA (Fig. 1A).Three hours after i.p. injection of mBSA, the macrophages haddisappeared but monocytes, neutrophils and eosinophils hademerged (Fig. 1A). Subsequently, at 24 h after i.p. injection ofmBSA, the number of neutrophils had decreased by 70%, butmacrophages were present again, probably having developedfrom the monocytes (Fig. 1A and B). Macrophages andeosinophils peaked in numbers at 2 days following i.p. injectionofmBSA and lymphocytes peaked at 5 days, atwhich time theywere the dominating cell type (Fig. 1A and B).

Please cite this article as: Tomasdottir, V., et al., Murine antigen-induand the adaptive phase of inflammation, J. Immunol. Methods (201

3.2. Neutrophils

The number of neutrophils peaked in the peritoneal cavity6 h after i.p. injection of mBSA but decreased bymore than 50%within 12 h (Fig. 1B). The neutrophils formed two distinctpopulations with the majority (~80%) belonging to a popula-tion of cells that were smaller and less granular (N1) thanneutrophils in the other subpopulation (N2) (Fig. 2A). Bothpopulations peaked at 6 h after administration ofmBSA, but theN2 population had disappeared from the peritoneal cavity by12 h (Fig. 2A). Neutrophils in the N2 population expressedmore CD11b, Ly6Gand slightlymore CXCR2 than neutrophils inthe N1 population (Fig. 2B).

3.3. Macrophages

Resident peritoneal macrophages had either low or highexpression of F4/80, with both populations expressing CD11b(Fig. 3A) and CD115 (data not shown). The F4/80high macro-phages constituted around 90% of total macrophages in theperitoneum prior to administration of mBSA. Following mBSAadministration, the resident macrophages almost disappearedfrom the peritoneal cavity but monocytes, which did notexpress F4/80 emerged. At 24 h following i.p. injection ofmBSA,F4/80high macrophages reappeared in the peritoneal cavity andpeaked at 48 h (Fig. 3A). They were smaller, less granular andexpressed less CD11b than the resident macrophages (Figs. 1Aand 3A). These macrophages had much higher expressionof the atypical chemokine receptor D6 and the chemokinereceptor CCR7 than resident peritoneal macrophages (Fig. 3B).Expression levels of D6 and CCR7, which are thought to be ofimportance in resolution of inflammation (Forster et al., 1999;McKimmie and Graham, 2006; Di Liberto et al., 2008), peakedat 2 days (Fig. 3C). F4/80low macrophages reappeared in theperitoneum 48 h after administration of mBSA (Fig. 3A). Theydid not express D6 or CCR7, but expressed the dendritic cell(DC) marker CD11c and the integral membrane protein CD138(syndecan-1) (Fig. 3B). Expression levels of CD138 werehighest at 10 days following mBSA administration (Fig. D).

3.4. Eosinophils

Eosinophils appeared in the peritoneal cavity 3 h afteradministration ofmBSA, peaked at 48 h and declined thereafter(Fig. 1A and B). When the number of eosinophils had reachedhalf of their maximum number, the number of neutrophils haddecreased by half and by the time the number of eosinophilspeaked the neutrophils had almost disappeared (Fig. 1B, insert).The eosinophils had a wide range of granularity (Fig. 1A), buttheir average granularity did not change throughout theexperiment (data not shown). Expression levels of the chemo-kine receptor CCR3 peaked at 48 h after administration ofmBSA,whereas mean expression levels of CD11b peaked at 6 h anddeclined rapidly thereafter (Fig. 4).

3.5. NK, T and B cells

NK (NK1.1+) cells appeared in the peritoneal cavity of mice3 h after i.p. injection of mBSA (Fig. 5A). They were scant at 3and 6 h, but subsequently their numbers increased dramaticallyand at 48 h they constituted around 2% of total peritoneal cells

ced inflammation—Amodel for studying induction, resolution4), http://dx.doi.org/10.1016/j.jim.2014.09.004

Page 4: Murine antigen-induced inflammation—A model for studying induction, resolution and the adaptive phase of inflammation

Fig. 1. Effects of mBSA administration on peritoneal cell populations. Mice were immunized twice with mBSA and injected 1 week later intraperitoneally with mBSA.Peritoneal fluid was collected at indicated time points. Cells were stained with fluorochrome-labeled monoclonal antibodies against surface molecules as listed in theMaterials and Methods section and analyzed by flow cytometry. Cell numbers were determined using Countess automated cell counter. (A) Dot plot with forward andside scatter analysis of peritoneal leukocytes prior to and at different time points following mBSA administration. Results are representative of 4 independentexperiments. (B) Number of neutrophils, monocytes, macrophages, eosinophils and lymphocytes prior to and at different time points following mBSA administration.Data are expressed as mean± SEM, n=4–7 per time point. Insert shows the number of the peritoneal cells during the first 24 h following induction of inflammation.*Different from 0 h; #different from previous time point; P b 0.05. L, lymphocytes (identified by expression of B220, CD90.2 or NK1.1); Mφ, macrophages (detected byexpression of F4/80 and CD11b); N, neutrophils (detected by expression of CD11b, Ly6G and CXCR2); E, eosinophils (detected by expression of CCR3); M, monocytes(detected by expression of CD115 and lack of expression of F4/80).

4 V. Tomasdottir et al. / Journal of Immunological Methods xxx (2014) xxx–xxx

(Fig. 5A). Lymphocytes constituted 42% of total peritoneal cellsprior to induction of peritonitis, but around 50% when theirnumbers peaked at 5 days (Fig. 1B). The number of T cells in theperitoneum increased after i.p. injection of mBSA, peaking at2 days (Fig. 5B) when they constituted around 20% oflymphocytes, with CD4+, CD8+ and CD4−CD8− cells compris-ing 43%, 23% and 34% of total T cells, respectively (Fig. 5B). B cellsconstituted around 40% of peritoneal lymphocytes prior tomBSA administration with B1 cells (B220lowIgD−) comprising75% and B2 cells (B220lowIgDhighCD5−) 25% of the total B cells(Fig. 5C). The number of B cells increased following mBSAadministration and increased again after a slight drop in theirnumbers at 24 h, peaking at 5 days (Fig. 5C). There were few B2cells in the peritoneal cavity prior to and early after mBSAadministration, but their numbers had increased at 2 and5 days. B1a (CD5+) cells constituted 65% and B1b (CD5−) cells35% of total B1 cells prior to mBSA administration (data notshown). Peritoneal T and B cells unregulated CD11b on theirsurface after mBSA administration. For B cells, the expressionlevels for CD11b were increased by 60% 24 h after administra-tion of mBSA, whereas for T cells, the increase was less (27%)and occurred later (48 h). The proportions of splenic T and Bcells were 29 and 58%, respectively, prior to induction of

Please cite this article as: Tomasdottir, V., et al., Murine antigen-induand the adaptive phase of inflammation, J. Immunol. Methods (201

inflammation and did not change during the time of peritonitis(data not shown).

3.6. IgM and IgG levels

Serum levels of mBSA-specific IgM antibodies increasedgradually and peaked 5 days after i.p. injection of mBSA(Fig. 6). Serum levels of mBSA-specific IgG antibodiestransiently decreased following administration of mBSA,most likely because of formation of IgG-mBSA immunecomplexes, but increased again and were similar to thatprior to mBSA administration at 10 days (Fig. 6).

3.7. Cytokines, chemokines and soluble cytokine receptors

Intraperitoneal administration of mBSA resulted in a rapidincrease in peritoneal concentrations of the pro-inflammatorycytokines and chemokines IL-1β, IL-6, IL-12, CCL2, CCL3 andCCL11, with peak levels occurring at 3 h, except for CCL11,which peaked at 6 h, and IL-12, which peaked at 12 h (Fig. 7Aand B). Peritoneal concentration of IL-1ra peaked 6 h afteradministration ofmBSA, sTNF-R at 24h,whereas concentrations

ced inflammation—Amodel for studying induction, resolution4), http://dx.doi.org/10.1016/j.jim.2014.09.004

Page 5: Murine antigen-induced inflammation—A model for studying induction, resolution and the adaptive phase of inflammation

Fig. 2. Effects of mBSA administration on N1 and N2 neutrophils. Mice were immunized twice with mBSA and injected 1 week later intraperitoneally with mBSA.Peritoneal fluid was collected at indicated time points. Cells were stained with fluorochrome-labeled monoclonal antibodies against CXCR2, Ly6G and CD11b andanalyzed by flow cytometry. Cell numberswere determined using Countess automated cell counter. (A) Number of N1 andN2 neutrophils prior to and at different timepoints following mBSA administration. Insert shows a dot plot with forward and side scatter analysis of neutrophils 6 h following mBSA administration. (B) Meanfluorescence of CD11b, Ly6G and CXCR2 on N1 (thick black line) and N2 (thin black line) neutrophils 6 h following administration of mBSA. Data are expressed asmean ± SEM, n = 4–7 per time point. *Different from 0 h; #different from previous time point; P b 0.05.

5V. Tomasdottir et al. / Journal of Immunological Methods xxx (2014) xxx–xxx

of sIL-6R and TGF-β1 did not peak until 2 days after peritonealadministration of mBSA (Fig. 7C and D).

4. Discussion

The resolution phase of acute self-limited inflammation hasbeen thoroughly described in murine zymosan-induced peri-tonitis (Bannenberg et al., 2005; Stables and Gilroy, 2011). Inthat model, the induction of acute inflammation is character-ized by a rapid increase in neutrophils with subsequent influxof macrophages and this is followed by resolution, character-ized by a marked decline in neutrophils and a change frominflammatory to resolution-type macrophages. This process isalso reflected in changes in immune mediators, which evolvefrom inflammatory cytokines to anti-inflammatory cytokinesand pro-resolution type mediators (Bannenberg et al., 2005;Stables and Gilroy, 2011). In autoimmune diseases, antigen-specific response is often responsible for the consistentlow-grade inflammation, which plays a role in pathogenesisof the disease. Since zymosan-induced peritonitis does not (atall) or minimally involve an antigen-specific immune response

Please cite this article as: Tomasdottir, V., et al., Murine antigen-induand the adaptive phase of inflammation, J. Immunol. Methods (201

(Rajakariar et al., 2008), it is important to also study inductionand resolution of inflammation in amurinemodel that involvesthe adaptive immune response and, thus, is mimicking a flare-up in autoimmunity. In the current study, we have examinedthe kinetics of cellular and immunemediator responses duringthe resolution phase of inflammation in murinemBSA-inducedacute peritonitis.

The induction phase of the antigen-induced inflammationin the present study was more rapid (Tmax = 6 h) but lesssevere (Ψmax = 3.3 × 106) and resolved more rapidly (Ri =5 h) than the zymosan-induced peritonitis (Tmax = 12 h;Ψmax = 15 × 106; Ri = 8 h) (Bannenberg et al., 2005). Twopopulations of neutrophils were observed in the present study,one population (N2), which included neutrophils that werelarger and more granulated than the majority of the neutro-phils in the other population (N1), and may have been in amore activated state, as they expressed more CD11b. We havepreviously described two circulating neutrophil populations inendotoxin-induced acute inflammation, but in that model,peritoneal neutrophils were a homogenous population(Arnardottir et al., 2012). Several other studies have shown

ced inflammation—Amodel for studying induction, resolution4), http://dx.doi.org/10.1016/j.jim.2014.09.004

Page 6: Murine antigen-induced inflammation—A model for studying induction, resolution and the adaptive phase of inflammation

Fig. 3. Effects of mBSA administration on peritoneal macrophages. Mice were immunized twice with mBSA and injected 1 week later intraperitoneally with mBSA.Peritoneal fluid was collected at indicated time points. Cells were stained with fluorochrome-labeled monoclonal antibodies against surface molecules as listed in theMaterials and Methods section and analyzed by flow cytometry. Circular gates are drawn around the different macrophage populations, expressing high or mediumlevels of F4/80 and at 5 days the F4/80highmacrophages are also divided intomacrophages expressingmediumand high levels of CD11b. (A) FACS analysis of peritonealleukocytes at different time points prior to and following peritoneal mBSA administration. (B) Mean fluorescence of D6, CCR7, CD11c and CD138 on F4/80high (thickblack line) and F4/80med (thin black line) macrophages 48 h following administration of mBSA, with isotypic control being depicted in light grey. (C) Mean expressionlevels of D6 and CCR7 on F4/80high macrophages at different time points following mBSA administration. (D) Mean expression levels of CD138 on F4/80med

macrophages at different time points, prior to and following mBSA administration. Data are expressed as mean ± SEM, n = 4–7 per time point. *Different from 0 h;#different from previous time point; P b 0.05. MFI, mean expression levels.

6 V. Tomasdottir et al. / Journal of Immunological Methods xxx (2014) xxx–xxx

heterogeneous neutrophil populations with different pheno-types and roles, changing during the different phases of theinflammatory process (reviewed in (Beyrau et al., 2012)).

The resolution phase of inflammation is not only character-ized by the disappearance of neutrophils from the inflammatory

Fig. 4. Effects of mBSA administration on CCR3 and CD11b expression on peritoneal eintraperitoneally withmBSA. Peritoneal fluid was collected at indicated time points. Ceand CD11b and analyzed by flow cytometry. Data are expressed as mean ± SEM, n =P b 0.05. MFI, mean expression levels.

Please cite this article as: Tomasdottir, V., et al., Murine antigen-induand the adaptive phase of inflammation, J. Immunol. Methods (201

exudate but also by the emergence of resolution phasemacrophages (Bystrom et al., 2008; Stables et al., 2011). In thepresent study, the two populations of macrophages emergingfollowing i.p. injection of mBSA, i.e., the F4/80high macrophages,appearing at 24 h and peaking at 48 h, and the F4/80low

osinophils. Mice were immunized twice with mBSA and injected 1 week laterlls were stainedwith fluorochrome-labeledmonoclonal antibodies against CCR34–7 per time point. *Different from 0 h; #different from previous time point;

ced inflammation—Amodel for studying induction, resolution4), http://dx.doi.org/10.1016/j.jim.2014.09.004

Page 7: Murine antigen-induced inflammation—A model for studying induction, resolution and the adaptive phase of inflammation

Fig. 5. Effects of mBSA administration on peritoneal lymphocytes. Mice wereimmunized twice with mBSA and injected 1 week later intraperitoneally withmBSA. Peritoneal fluidwas collected at indicated time points. Cellswere stainedwith fluorochrome-labeledmonoclonal antibodies against surfacemolecules aslisted in the Materials and Methods section and analyzed by flow cytometry.(A) Number of NK cells prior to and at different time points following mBSAadministration. (B) Number of T cells prior to and at different time pointsfollowing mBSA administration. (C) Number of B cells prior to and at differenttime points following mBSA administration. Data are expressed as mean ±SEM, n=4–7 per time point. *Different from 0 h; #different from previous timepoint; P b 0.05.

7V. Tomasdottir et al. / Journal of Immunological Methods xxx (2014) xxx–xxx

macrophages, appearing at 48 h, both expressed high levels ofmolecules linked with resolution. The F4/80high macrophagesexpressed high levels of the chemokine receptor CCR7 and theatypical chemokine receptor D6. CCR7 facilitates leukocytemigration to regional lymph nodes (Forster et al., 1999);hence, the high expression of this chemokine receptor onmacrophages present in the peritoneal cavity at 48 h in thepresent study may indicate a high potential of these macro-phages to migrate to regional lymph nodes. D6 is a decoy

Please cite this article as: Tomasdottir, V., et al., Murine antigen-induand the adaptive phase of inflammation, J. Immunol. Methods (201

receptor that binds most inflammatory CC chemokines and hasbeen suggested to play a role in resolution of CC-chemokine-driven inflammatory responses (Jamieson et al., 2005). Supportfor a role for D6 in resolution comes from studies showing thatD6-deficient mice have exaggerated inflammatory response(Jamieson et al., 2005; Di Liberto et al., 2008; Graham, 2009;Vetrano et al., 2010).

The F4/80high macrophages present in the peritoneal cavity48 h after induction of inflammation in the present study hadlower expression of CD11b than the resident peritonealmacrophages and may therefore be similar to the pro-resolving CD11blow macrophages that were recently reportedto emerge in vivo during resolution of zymosan-inducedperitonitis (Schif-Zuck et al., 2011). In that study, macrophagesconverted from a CD11bhigh to a CD11blow phenotype uponinteractionwith apoptotic cells ex vivo (Schif-Zuck et al., 2011).Recognition of apoptotic cells by macrophages has also beenshown to evoke signaling events that block the release of pro-inflammatory mediators from macrophages (Patel et al., 2006)and induce the production of TGF-β, resulting in acceleratedresolution of inflammation (Huynh et al., 2002; Mitchell et al.,2002). In the present study, TGF-β concentration peakedsimultaneously with the number of F4/80high macrophages,indicating active resolution of inflammation at 48 h afterinduction of inflammation in the antigen-induced peritonitis.

Although the F4/80low macrophages expressed the DCmarker CD11c, their expression of F4/80 and lack of CCR7expression indicates that they are not typical antigen-presenting DCs (Dieu et al., 1998). These cells also expressedCD138, a surface heparin sulfate proteoglycan that has beenshown to bind to and modulate the activity of chemokines,selectins, integrins and other adhesionmolecules (Gotte, 2003;Gotte and Echtermeyer, 2003; Bartlett et al., 2007). CD138 hasbeen proposed to play a role in resolution of inflammation byinhibiting leukocyte adhesion, reducing or inhibiting expres-sion of pro-inflammatory factors or by removing inflammatorychemokines (Teng et al., 2012). Expression of CD138 on theF4/80med macrophages was highest 5 and 10 days afterinduction of inflammation indicating that these macrophagesare regulatory and may play a role in return to homeostasis.

In the present study, a considerable influx of eosinophils intothe peritoneum was observed, already detectable 3 h after theinitiation of inflammation and peaking at 48 h. The presence ofeosinophils in an inflammatory exudate is conventionallybelieved to signify unwanted allergic type hypersensitivityreaction. This is unlikely to be the case in the mBSA-inducedinflammation as the eosinophils emerge in a CFA-induced, Th1-polarized environment and BSA-specific IgE antibodies werehardly detectable in the serum of these mice at any time point(data not shown). In allergic eosinophilic reactions, such as inasthma, there is commonly increased CD11b expression oneosinophils (Walker et al., 1993; Chiba et al., 2005), but themean expression levels of CD11b on peritoneal eosinophils inthe present study was decreased at the time when theirnumbers peaked. It is also unlikely that the eosinophils have acytotoxic function or release cytotoxic granules in thesemice, astheir mean granularity did not change throughout the study.

Several observations in the present study support the viewthat the influx of eosinophilsmay have an important role in theresolution process. First, at the same time as the number ofeosinophils increased in the peritoneum, therewas a reciprocal

ced inflammation—Amodel for studying induction, resolution4), http://dx.doi.org/10.1016/j.jim.2014.09.004

Page 8: Murine antigen-induced inflammation—A model for studying induction, resolution and the adaptive phase of inflammation

Fig. 6.Effects ofmBSAadministration on serum levels ofmBSA-specific IgMand IgG.Micewere immunized twicewithmBSA and injected 1week later intraperitoneallywith mBSA. Serum was collected at indicated time points and concentrations of BSA-specific IgM and IgG antibodies measured by ELISA. Values are in arbitrary units(AU). Data are expressed as mean ± SEM, n = 4–7 per time point. *Different from 0 h; #different from previous time point; P b 0.05.

8 V. Tomasdottir et al. / Journal of Immunological Methods xxx (2014) xxx–xxx

decrease in the number of neutrophils, similar to that observedby (Yamada et al., 2011) using the zymosan-induced peritoni-tis model and demonstrating a resolution promoting role forthe eosinophils. Second, the eosinophils peaked in numbers atthe same time as the resolution type F4/80high macrophagesexpressing D6 and CCR7. Third, the levels of the anti-inflammatory/immunosuppressive molecules TGF-β and sIL-6R reached maximum as the eosinophils peaked in numbers.The TGF-β may have been produced by the peritonealeosinophils as eosinophils have been shown to be a major

Fig. 7. Effects of mBSA administration on peritoneal concentrations of cytokines, chemand injected 1 week later intraperitoneally with mBSA. Peritoneal fluid was collected acytokine receptorsmeasured by ELISA. (A) IL-1β, IL-6 and IL-12. (B) CCL2, CCL3 and CCLas mean ± SEM, n = 4–7 per time point. *Different from 0 h; #different from previous

Please cite this article as: Tomasdottir, V., et al., Murine antigen-induand the adaptive phase of inflammation, J. Immunol. Methods (201

source of this immunosuppressive cytokine (Kadin et al.,1993).

A role for NK cells in resolution of inflammation has recentlybeen suggested as they induce neutrophil apoptosis in vitro(Thoren et al., 2012) and are essential for effective resolution ofallergic lung inflammation in vivo (Haworth et al., 2011). Thekinetics of the NK response in the current study differed fromthat in the allergic lung inflammation study as the number ofNK cells peaked earlier (24 h vs 48 h), and the increase in NKcell number wasmuch greater in the current study (more than

okines and soluble cytokine receptors. Mice were immunized twice with mBSAt indicated time points and concentrations of cytokines, chemokines and soluble11 (eotaxin 1). (C) sIL-6R and sTNF-R1. (D) IL-1ra and TGF-β. Data are expressedtime point; P b 0.05.

ced inflammation—Amodel for studying induction, resolution4), http://dx.doi.org/10.1016/j.jim.2014.09.004

Page 9: Murine antigen-induced inflammation—A model for studying induction, resolution and the adaptive phase of inflammation

9V. Tomasdottir et al. / Journal of Immunological Methods xxx (2014) xxx–xxx

thirteen fold) (Haworth et al., 2011). In the present study, thenumber of NK cells peaked at the same time as the eosinophilsand the resolving macrophages, suggesting that they may playa role in the resolution of inflammation.

The early infiltration of both T and B cells into the peritonealcavity of the mice following induction of inflammation by i.p.injection of mBSA indicates involvement of the adaptiveimmune system in the antigen-induced response and thepresence of memory lymphocytes. The number of T cellspeaked earlier (48 h) than the number of B cells (5 days),perhaps reflecting the need of T cell-help for B cell activation.Similar to what has been reported for the mBSA-induced andzymosan-induced peritonitis (Cook et al., 2003), themajority ofthe peritoneal B cells, prior to and at all time points during theinflammation, were B1 cells. Although B1 cells are known toproduce high levels of polyspecific IgM antibodies, both duringhomeostasis and infections, B1b cells are able to secreteantigen-specific antibodies following stimulation via the B cellreceptor (Foote and Kearney, 2009). Therefore, the B1b cells inthe present study are most likely the producers of the BSA-specific IgM antibodies observed in the serum.

The immunization with mBSA resulted in high levels ofBSA-specific IgG antibodies in serum of the mice prior toinduction of peritonitis. Upon induction of peritonitis, the IgGlevels dropped temporarily, undoubtedly because of immunecomplex formation between the IgG antibodies and the mBSA.These immune complexes are most likely the inducers of theperitonitis in the mice.

It has been demonstrated that resolution of inflammation isinitiated within 6 hours in models of acute self-limited inflam-matory response to carrageenan and zymosan (Bannenberget al., 2005). In the current study, the early rise in theconcentration of the anti-inflammatorymediator IL-1ra, peakingat 6 h, indicates early induction of resolution of inflammation inthe antigen-induced peritonitis model. Other immunosuppres-sive molecules that were analyzed, sTNF-R, sIL-6R and TGF-β,peaked at 24–48 h, the time at which neutrophils weredisappearing from the peritoneal cavity, and may have beeninvolved in reducing their numbers as they have previously beenassociated with reducing neutrophil accumulation in peritonealinflammation (Hurst et al., 2001).

In summary, we have delineated cellular and molecularresponses in the induction, resolution and adaptive immunephases of the mBSA-induced peritonitis model. Our studysuggests that numerous cell types play an active role in theresolution and adaptive phases of the immune response,including neutrophils, resolution-type macrophages, eosino-phils, NK cells and B and T lymphocytes. Our results indicatethat themBSA-induced peritonitis model is an excellent modelto study resolution of inflammation and the role of the adaptiveimmune responses in later stages of inflammation. We havepreviously used thismodel to demonstrate the ability of dietaryomega-3 polyunsaturated fatty acids to affect the induction,resolution and the adaptive immune phases of the inflamma-tion (Tomasdottir et al., 2013, 2014).

Acknowledgments

This project was supported by grants from the IcelandicResearch Fund, the University of Iceland Research Fund and theLandspitali University Hospital Research Fund.

Please cite this article as: Tomasdottir, V., et al., Murine antigen-induand the adaptive phase of inflammation, J. Immunol. Methods (201

References

Ariel, A., Timor, O., 2013. Hanging in thebalance: endogenous anti-inflammatorymechanisms in tissue repair and fibrosis. J. Pathol. 229, 250.

Arnardottir, H.H., Freysdottir, J., Hardardottir, I., 2012. Two circulating neutrophilpopulations in acute inflammation in mice. Inflamm. Res. 61, 931.

Bannenberg, G.L., Chiang, N., Ariel, A., Arita, M., Tjonahen, E., Gotlinger, K.H.,Hong, S., Serhan, C.N., 2005. Molecular circuits of resolution: formation andactions of resolvins and protectins. J. Immunol. 174, 4345.

Bartlett, A.H., Hayashida, K., Park, P.W., 2007. Molecular and cellular mecha-nisms of syndecans in tissue injury and inflammation. Mol. Cells 24, 153.

Beyrau, M., Bodkin, J.V., Nourshargh, S., 2012. Neutrophil heterogeneity inhealth and disease: a revitalized avenue in inflammation and immunity.Open Biol. 2, 120134.

Bystrom, J., Evans, I., Newson, J., Stables, M., Toor, I., van Rooijen, N., Crawford,M., Colville-Nash, P., Farrow, S., Gilroy, D.W., 2008. Resolution-phasemacrophages possess a unique inflammatory phenotype that is controlledby cAMP. Blood 112, 4117.

Chiba, T., Kamada, Y., Saito, N., Oyamada, H., Ueki, S., Kobayashi, Y., Kayaba, H.,Chihara, J., 2005. RANTES and eotaxin enhance CD11b and CD18 expressionon eosinophils from allergic patients with eosinophilia in the application ofwhole Blood flow cytometry analysis. Int. Arch. Allergy Immunol. 137(Suppl. 1), 12.

Cook, A.D., Braine, E.L., Hamilton, J.A., 2003. The phenotype of inflammatorymacrophages is stimulus dependent: implications for the nature of theinflammatory response. J. Immunol. 171, 4816.

Cook, A.D., Braine, E.L., Hamilton, J.A., 2004. Stimulus-dependent requirementfor granulocyte-macrophage colony-stimulating factor in inflammation. J.Immunol. 173, 4643.

Di Liberto, D., Locati,M., Caccamo, N., Vecchi, A., Meraviglia, S., Salerno, A., Sireci,G., Nebuloni,M., Caceres, N., Cardona, P.J., Dieli, F., Mantovani, A., 2008. Roleof the chemokine decoy receptor D6 in balancing inflammation, immuneactivation, and antimicrobial resistance in Mycobacterium tuberculosisinfection. J. Exp. Med. 205, 2075.

Dieu, M.C., Vanbervliet, B., Vicari, A., Bridon, J.M., Oldham, E., Ait-Yahia, S.,Briere, F., Zlotnik, A., Lebecque, S., Caux, C., 1998. Selective recruitment ofimmature and mature dendritic cells by distinct chemokines expressed indifferent anatomic sites. J. Exp. Med. 188, 373.

Foote, J.B., Kearney, J.F., 2009. Generation of B cell memory to the bacterialpolysaccharide alpha-1,3 dextran. J. Immunol. 183, 6359.

Forster, R., Schubel, A., Breitfeld, D., Kremmer, E., Renner-Muller, I., Wolf, E.,Lipp, M., 1999. CCR7 coordinates the primary immune response byestablishing functional microenvironments in secondary lymphoid organs.Cell 99, 23.

Gilroy, D.W., Lawrence, T., Perretti,M., Rossi, A.G., 2004. Inflammatory resolution:new opportunities for drug discovery. Nat. Rev. Drug Discov. 3, 401.

Gotte, M., 2003. Syndecans in inflammation. FASEB J. 17, 575.Gotte, M., Echtermeyer, F., 2003. Syndecan-1 as a regulator of chemokine

function. ScientificWorldJournal 3, 1327.Graham, G.J., 2009. D6 and the atypical chemokine receptor family: novel

regulators of immune and inflammatory processes. Eur. J. Immunol. 39,342.

Haworth, O., Cernadas, M., Levy, B.D., 2011. NK cells are effectors for resolvin E1in the timely resolution of allergic airway inflammation. J. Immunol. 186,6129.

Hurst, S.M., Wilkinson, T.S., McLoughlin, R.M., Jones, S., Horiuchi, S., Yamamoto,N., Rose-John, S., Fuller, G.M., Topley, N., Jones, S.A., 2001. Il-6 and its solublereceptor orchestrate a temporal switch in the pattern of leukocyterecruitment seen during acute inflammation. Immunity 14, 705.

Huynh, M.L., Fadok, V.A., Henson, P.M., 2002. Phosphatidylserine-dependentingestion of apoptotic cells promotes TGF-beta1 secretion and theresolution of inflammation. J. Clin. Invest. 109, 41.

Jamieson, T., Cook, D.N., Nibbs, R.J., Rot, A., Nixon, C., McLean, P., Alcami, A., Lira,S.A., Wiekowski, M., Graham, G.J., 2005. The chemokine receptor D6 limitsthe inflammatory response in vivo. Nat. Immunol. 6, 403.

Kadin, M., Butmarc, J., Elovic, A., Wong, D., 1993. Eosinophils are the majorsource of transforming growth factor-beta 1 in nodular sclerosingHodgkin's disease. Am. J. Pathol. 142, 11.

Kolaczkowska, E., Barteczko,M., Plytycz, B., Arnold, B., 2008. Role of lymphocytesin the course of murine zymosan-induced peritonitis. Inflamm. Res. 57, 272.

McKimmie, C.S., Graham, G.J., 2006. Leucocyte expression of the chemokinescavenger D6. Biochem. Soc. Trans. 34, 1002.

Mitchell, S., Thomas, G., Harvey, K., Cottell, D., Reville, K., Berlasconi, G., Petasis,N.A., Erwig, L., Rees, A.J., Savill, J., Brady, H.R., Godson, C., 2002. Lipoxins,aspirin-triggered epi-lipoxins, lipoxin stable analogues, and the resolutionof inflammation: stimulation of macrophage phagocytosis of apoptoticneutrophils in vivo. J. Am. Soc. Nephrol. 13, 2497.

Patel, V.A., Longacre, A., Hsiao, K., Fan, H., Meng, F., Mitchell, J.E., Rauch, J., Ucker,D.S., Levine, J.S., 2006. Apoptotic cells, at all stages of the death process,trigger characteristic signaling events that are divergent from and

ced inflammation—Amodel for studying induction, resolution4), http://dx.doi.org/10.1016/j.jim.2014.09.004

Page 10: Murine antigen-induced inflammation—A model for studying induction, resolution and the adaptive phase of inflammation

10 V. Tomasdottir et al. / Journal of Immunological Methods xxx (2014) xxx–xxx

dominant over those triggered by necrotic cells: implications for thedelayed clearance model of autoimmunity. J. Biol. Chem. 281, 4663.

Rajakariar, R., Lawrence, T., Bystrom, J., Hilliard, M., Colville-Nash, P., Bellingan,G., Fitzgerald, D., Yaqoob, M.M., Gilroy, D.W., 2008. Novel biphasic role forlymphocytes revealed during resolving inflammation. Blood 111, 4184.

Rogerio, A.P., Haworth, O., Croze, R., Oh, S.F., Uddin, M., Carlo, T., Pfeffer, M.A.,Priluck, R., Serhan, C.N., Levy, B.D., 2012. Resolvin d1 and aspirin-triggeredresolvin d1 promote resolution of allergic airways responses. J. Immunol.189, 1983.

Schif-Zuck, S., Gross, N., Assi, S., Rostoker, R., Serhan, C.N., Ariel, A., 2011.Saturated-efferocytosis generates pro-resolving CD11b low macrophages:modulation by resolvins and glucocorticoids. Eur. J. Immunol. 41, 366.

Serhan, C.N., 2007. Resolution phase of inflammation: novel endogenous anti-inflammatory and proresolving lipid mediators and pathways. Annu. Rev.Immunol. 25, 101.

Stables, M.J., Gilroy, D.W., 2011. Old and new generation lipid mediators inacute inflammation and resolution. Prog. Lipid Res. 50, 35.

Stables, M.J., Shah, S., Camon, E.B., Lovering, R.C., Newson, J., Bystrom, J., Farrow,S., Gilroy, D.W., 2011. Transcriptomic analyses of murine resolution-phasemacrophages. Blood 118, e192.

Teng, Y.H., Aquino, R.S., Park, P.W., 2012. Molecular functions of syndecan-1 indisease. Matrix Biol. 31, 3.

Please cite this article as: Tomasdottir, V., et al., Murine antigen-induand the adaptive phase of inflammation, J. Immunol. Methods (201

Thoren, F.B., Riise, R.E., Ousback, J., Della Chiesa, M., Alsterholm, M., Marcenaro,E., Pesce, S., Prato, C., Cantoni, C., Bylund, J., Moretta, L., Moretta, A., 2012.Human NK Cells induce neutrophil apoptosis via an NKp46- and Fas-dependent mechanism. J. Immunol. 188, 1668.

Tomasdottir, V., Vikingsson, A., Freysdottir, J., Hardardottir, I., 2013. Dietary fishoil reduces the acute inflammatory response and enhances resolution ofantigen-induced peritonitis. J. Nutr. Biochem. 24, 1758.

Tomasdottir, V., Thorleifsdottir, S., Vikingsson, A., Hardardottir, I., Freysdottir, J.,2014. Dietary omega-3 fatty acids enhance the B1 but not the B2 cellimmune response in mice with antigen-induced peritonitis. J. Nutr.Biochem. 25, 111.

Vetrano, S., Borroni, E.M., Sarukhan, A., Savino, B., Bonecchi, R., Correale, C.,Arena, V., Fantini, M., Roncalli, M., Malesci, A., Mantovani, A., Locati, M.,Danese, S., 2010. The lymphatic system controls intestinal inflammationand inflammation-associated Colon Cancer through the chemokine decoyreceptor D6. Gut 59, 197.

Walker, C., Rihs, S., Braun, R.K., Betz, S., Bruijnzeel, P.L., 1993. Increasedexpression of CD11b and functional changes in eosinophils after migrationacross endothelial cell monolayers. J. Immunol. 150, 4061.

Yamada, T., Tani, Y., Nakanishi, H., Taguchi, R., Arita, M., Arai, H., 2011.Eosinophils promote resolution of acute peritonitis by producingproresolving mediators in mice. FASEB J. 25, 561.

ced inflammation—Amodel for studying induction, resolution4), http://dx.doi.org/10.1016/j.jim.2014.09.004