Skip to main content
Erschienen in: BMC Immunology 1/2015

Open Access 01.12.2015 | Research article

Dendritic cell immunoreceptor 1 alters neutrophil responses in the development of experimental colitis

verfasst von: Sumika Tokieda, Marie Komori, Toshifumi Ishiguro, Yoichiro Iwakura, Kazuhiko Takahara, Kayo Inaba

Erschienen in: BMC Immunology | Ausgabe 1/2015

Abstract

Background

Ulcerative colitis, an inflammatory bowel disease, is associated with the massive infiltration of neutrophils. Although the initial infiltration of neutrophils is beneficial for killing bacteria, it is presumed that persistent infiltration causes tissue damage by releasing antibacterial products as well as inflammatory cytokines. A murine C-type lectin receptor, dendritic cell immunoreceptor 1 (Dcir1), is expressed on CD11b+ myeloid cells, such as macrophages, dendritic cells and neutrophils. It was reported that Dcir1 is required to maintain homeostasis of the immune system to prevent autoimmunity, but it is also involved in the development of infectious disease resulting in the enhanced severity of cerebral malaria. However, the role of Dcir1 in intestinal immune responses during colitis remains unclear. In this study, we investigated the role of Dcir1 in intestinal inflammation using an experimental colitis model induced with dextran sodium sulfate (DSS).

Results

In contrast to wild type (WT) mice, Dcir1 −/− mice exhibited mild body weight loss during the course of DSS colitis accompanied by reduced colonic inflammation. Dcir1 deficiency caused a reduced accumulation of neutrophils in the inflamed colon on day 5 of DSS colitis compared with WT mice. Consistently, the production of a neutrophil-attracting chemokine, MIP-2, was also decreased in the Dcir1 −/− colon compared with the WT colon on day 5. There were fewer myeloperoxidase-positive neutrophils in the inflamed colon of Dcir1 −/− mice than in that of WT mice. Moreover, bone marrow neutrophils from Dcir1 −/− mice produced less reactive oxygen species (ROS) by lipopolysaccharide stimulation than those from WT mice. This suggests that Dcir1 deficiency decreases the accumulation of tissue destructive neutrophils during DSS colitis.

Conclusion

Dcir1 enhances the pathogenesis of DSS colitis by altering neutrophil recruitment and their functions.
Hinweise

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

ST, KT, and KI designed the research, ST, MK and TI conducted the research and analyzed the data. ST and MK performed statistical analysis. ST, KI and KT wrote the paper and have primary responsibility for its all content. YI provided essential mouse strains, valuable advice and technical expertise for these studies. All authors read and approved the final manuscript.
Abkürzungen
IBD
Inflammatory bowel disease
UC
Ulcerative colitis
ROS
Reactive oxygen species
MPO
Myeloperoxidase
DC
Dendritic cell
DCIR
DC immunoreceptor
ITIM
Immunoreceptor tyrosine–based inhibitory motif
SHP
Src homology 2 domain tyrosine phosphatase
DSS
Dextran sulfate sodium
MIP-2
Macrophage inflammatory protein-2

Background

Inflammatory bowel disease (IBD) is an inflammatory disorder of the gastrointestinal tract and is clinically subdivided into two forms: ulcerative colitis (UC) and Crohn’s disease [1]. The disease is characterized by inflammation of the intestine, and the clinical symptoms include weight loss, diarrhea accompanied by blood, and abdominal pain [2, 3]. The affected areas typically show transmural inflammation characterized by lymphoid hyperplasia, submucosal edema, ulcerative lesions and fibrosis [2]. The etiology of IBD still remains largely unclear. In recent years, however, epidemiologic and genetic studies in human and IBD-related animal models have suggested that genetic susceptibility factors affecting the host immune system and environmental factors such as gut microbiota contribute to the initiation and chronicity of IBD [46].
Neutrophils produce massive amounts of reactive oxygen species (ROS) that effectively destroy pathogens [7] and numerous antibacterial intracellular granules that contain myeloperoxidase (MPO) among other molecules [8]. Hypochlorous acid and other chlorinated oxidants generated by a catalytic reaction between chloride and hydrogen peroxide by MPO result in tissue injury under inflammatory conditions and protect from pathogenic infection [810]. In the gut of IBD patients, the excessive recruitment and accumulation of activated neutrophils were observed in association with mucosal injury and debilitating disease symptoms [7]. IBD can be accelerated by innate immune cells, which recognize pathogen-associated and damage-associated molecular patterns through pattern recognition receptors (PRRs) [11, 12].
Dendritic cell (DC) immunoreceptor (DCIR), a C-type lectin receptor, is a PRR and is expressed on DCs, monocytes, macrophages, B cells, and neutrophils in humans [13, 14]. The cytoplasmic tail of DCIR contains an immunoreceptor tyrosine-based inhibitory motif (ITIM), which transduces immunoregulatory signals via interactions with Src homology 2 domain tyrosine phosphatase (SHP)-1 and SHP-2 [14]. Similar to other C-type lectin receptors such as Mincle, which is expressed on macrophages and recognizes pathogen-associated molecular patterns and damage-associated molecular patterns to initiate an immune response [15], DCIR can bind to endogenous carbohydrates as well as pathogenic and viral antigens [16, 17].
In mice, Dcir consists of four homologs (Dcir1 to 4). Although Dcir1 and Dcir2 contain ITIM sequences, only Dcir1 has been shown to interact with SHP-1 and SHP-2 [14, 18]. Several studies showed that murine Dcir1 is also expressed on CD11b+ myeloid cells, including monocytes, macrophages, and DCs; however, Dcir2 is mainly expressed on CD8 DCs [14, 16, 19]. We recently reported that a lack of Dcir1 exacerbated autoimmune arthritis caused by the spontaneous expansion and activation of DCs [20]. Other studies demonstrated that Dcir1 was essential for the development of cerebral malaria and that Dcir1 deficiency reduced brain inflammation accompanied with decreased tumor necrosis factor (TNF)-α production and modulated the activation of T cells [21]. These results suggest that Dcir1 is involved in disease symptoms both positively and negatively.
Infiltration by neutrophils and monocytes is a characteristic histologic feature of lesions of active UC [22]. Neutrophils and Ly6Chi monocytes are considered to promote intestinal inflammation during acute dextran sulfate sodium (DSS) induced colitis (DSS colitis) [23, 24]; therefore, we hypothesized that Dcir1 may have an effect on the symptoms of colitis by altering the behavior and functions of these cells.
This study examined the effect of Dcir1 deficiency in intestinal inflammation using a DSS colitis model. We found that Dcir1 −/− mice developed milder colitis with low accumulation of activated neutrophils and expression of macrophage inflammatory protein-2 (MIP-2) in the colon than WT mice. Neutrophils from Dcir1 −/− bone marrow also had less ROS production in response to lipopolysaccharide (LPS) than those from WT mice. In conclusion, our data suggest that Dcir1 is responsible for the development of acute colitis by promoting neutrophil recruitment and altering neutrophil functions.

Methods

Mice

C57BL/6 mice were purchased from Japan SLC (Hamamatsu, Japan). Dcir1-deficient (Dcir1 −/− ) mice on a C57BL/6 background [20] were obtained from Dr. Iwakura (Tokyo University of Science, Chiba, Japan). All mice used in this study were female and aged between 8 and 10 weeks. Dcir1 −/− or C57BL/6 wild type (WT) control mice were co-housed after weaning until use. All experiments were performed according to the Institutional Guidelines for Animal Use and Experimentation of Kyoto University (Permit Number: Lif-K13020).

DSS colitis

Dcir1 −/− or WT mice were provided with 2.0 % (wt/vol) DSS (reagent grade DSS salt; molecular mass = 36–50 kDa) (MP Biomedicals, Irvine, CA) dissolved in drinking water for 5 days, followed by 3 days with normal water. Mice given normal water for the entire period were used as controls. Individual mouse body weight was recorded daily for each group. On day 8, the length of the cecum and colon was measured after dissection by placing them on a paper towel.

Histological analysis

After washing in phosphate buffered saline (PBS), the cecum and colon were cut longitudinally and fixed with Bouin’s solution (71.4 % saturated picric acid, 23.8 % formaldehyde, and 4.8 % acetic acid) for 2 h at room temperature and embedded in paraffin. Two sections (4–6 μm thick) from a remote area of the cecum, and sections from the descending, transverse, and ascending colon from individual mice were stained with hematoxylin-eosin (HE) and observed using a BZ-8000 Biozero imaging device (Keyence, Osaka, Japan).
Histological colitis scores were determined as previously described [25]. Briefly, subscores were as follows: ulceration: 0, no ulcers; 1, one ulcer; 2, two ulcers; 3, three ulcers; and 4, > 3 ulcers: epithelium: 0, normal morphology; 1, loss of goblet cells; 2, loss of goblet cells in large areas; 3, loss of crypts; and 4, loss of crypts in large areas (>50 %) and/or foci of polypoid regeneration: infiltration: 0, no infiltrate; 1, infiltrate around crypt bases; 2, infiltrate reaching to lamina muscularis mucosae; 3, extensive infiltration reaching lamina muscularis mucosae, thickening of the mucosa with abundant edema; and 4, infiltration of the lamina submucosa: lymphoid follicles: 0, no lymphoid follicles; 1, one lymphoid follicle; 2, two lymphoid follicles; 3, three lymphoid follicles; and 4, > 3 lymphoid follicles. The colitis score of each mouse represents the mean of the different histological subscores.
For the immunohistological detection of neutrophils, 10-μm-thick frozen sections from DSS-treated WT and Dcir1 −/− colonic tissues were fixed with 4 % paraformaldehyde, and stained with rabbit anti-MPO polyclonal antibody (Ab) (Abcam, Cambridge, MA) followed by incubation with donkey-anti rabbit IgG-Cy3 (BioLegend, San Diego, CA) and then with anti-Ly6G-FITC (BD Biosciences, San Jose, CA), anti-CD11b-APC (eBioscience, San Diego, CA) and DAPI (Wako Pure Chemical Industries, Ltd., Osaka, Japan). After refixation with 1 % paraformaldehyde for 10 min, the specimens were mounted with Vectashield mounting medium (Vector Laboratories, Burlingame, CA) and observed using a deconvolution microscope BX51-FL (Olympus, Tokyo, Japan). MPO+CD11b+Ly6G+ cells were counted as activated neutrophils. Data represent number of neutrophils/whole area of a vertical section.

Isolation of mononuclear cells

Isolation of lamina propria mononuclear cells (LPMCs) was performed as described previously [26] with some modifications. Briefly, colons were opened longitudinally and washed to remove fecal content by shaking in Hank’s Balanced Salt Solution containing 5 mM EDTA for 20 min at 37 °C, followed by the removal of epithelial cells and fat tissue. After cutting the colon into small pieces, tissues were incubated with RPMI 1640 containing 4 % fetal bovine serum (FBS), 1 mg/ml collagenase D (Roche, Basel, Switzerland), 0.5 mg/ml dispase (Gibco, Carlsbad, CA) and 40 μg/ml DNase (Sigma–Aldrich, St. Louis, MO) for 50 min at 37 °C in a shaking water bath. The digested tissues were washed with Hank’s Balanced Salt Solution containing 5 mM EDTA, resuspended in 4 ml of 40 % Percoll (GE Healthcare, Tokyo, Japan) and overlaid on 2 ml of 80 % Percoll in a 15 ml conical tube. Percoll gradient separation was performed by centrifugation at 1200 × g for 20 min at 25 °C. The cells at the interface were collected and used as LPMCs.

Flow cytometry

LPMCs suspended in staining buffer (PBS containing 1 % FBS, 5 mM EDTA and 0.02 % NaN3) were incubated with anti-CD16/32 monoclonal antibodies (mAb) for 30 min at 4 °C to block nonspecific binding. Then, cells were stained for 30 min at 4 °C with a combination of the following fluorescent mAbs: FITC-conjugated anti-Ly6G or anti-DX5 (BD Biosciences), PE-conjugated anti-CD11c (BD Biosciences), PE-Cy7-conjugated anti-CD11b (BD Biosciences), Alexa Fluor 647-conjugated anti-Ly6C (BioLegend), APC-conjugated anti-CD3ε (eBioscience) or anti-CD19 (eBioscience). Dead cells were gated out by staining with 7-aminoactinomycin D (7-AAD) (Life Technologies, Carlsbad, CA). Stained cells were acquired using FACSCalibur (BD Biosciences) or FACSAriaIII (BD Biosciences), and data were analyzed with FlowJo software (TreeStar Inc., Ashland, OR).

Cytokine measurement

After treatment with DSS for 5 days, ceca and colons were obtained from Dcir1 −/− or WT mice. Colons were divided into three pieces and ceca were washed and weighed. Each segment was cultured in RPMI 1640 medium for 24 h, and cytokines, interleukin (IL)-1β, IL-6, IL-10, IL-12, TNF-α and monocyte chemoattractant protein-1 (MCP-1), in the medium were measured using a cytometric bead array (BD Biosciences) according to the manufacturer’s instructions. Data were analyzed with FCAP Array software (BD Biosciences). Data were analyzed by FCAP Array software (BD Biosciences). The concentrations of MIP-2 and KC were measured by ELISA (PeproTech, Rocky Hill, NJ) according to the manufacturer’s instructions.

Migration assay of neutrophils

Chemotactic activity of neutrophils was assessed using 3-μm pore size transwells (Costar, Washington, DC). After removing red blood cells with ACK lysis buffer (0.15 M NH4Cl, 10 mM KHCO3, 0.1 mM Na2EDTA, pH 7.2), bone marrow cells were blocked with anti-CD16/32 mAb, and stained with FITC-anti-Ly6G and PE-anti-CD11b mAbs (eBioscience). Then, cell suspensions (106/well) in RPMI 1640 containing 0.1 % bovine serum albumin in the upper well were placed above a lower well with or without 100 ng of MIP-2 (PeproTech). One hour later, input cells and cells that had migrated into the lower well were counted and analyzed by flow cytometry.

Neutrophil preparation and measurement of oxidative burst

Ly6G+ CD11b+ cells in bone marrow were sorted as neutrophils using FACSAriaIII. Purified neutrophils were stimulated in the presence or absence of 100 ng/ml Ultra-pure LPS from E. coli O111:B4 (InvivoGen, San Diego, CA) or 10 ng/ml granulocyte-macrophage colony-stimulating factor (GM-CSF) (Kirin Brewery, Gunma, Japan) for 6 h, and then incubated with 10 μM of dihydrorhodamine-123 (DHR-123) (Sigma–Aldrich) for 30 min in quadruple cultures. Fluorescence intensity was determined using FACSCalibur. Results were calculated according to the following formula: rhodamin-123 fluorescence = mean fluorescence intensity (MFI) of stimulated cells/MFI of unstimulated cells × 100.

Statistical analysis

Differences between Dcir1 −/− and WT groups or treated and nontreated groups were evaluated using the unpaired Student’s t-test. A P-value of < 0.05 was considered statistically significant.

Results

Milder symptoms of DSS colitis in Dcir1 −/− compared with WT mice

To elucidate the roles of Dcir1 in acute colitis, Dcir1 −/− and WT mice were treated with drinking water containing 2.0 % DSS for 5 days, followed by normal water for 3 days. DSS-treated WT and Dcir1 −/− mice started losing body weight on day 4 and day 5, respectively, and both groups showed a gradual loss of weight thereafter (Fig. 1a). Weight loss in DSS-treated WT mice was significantly greater than that in Dcir1 −/− mice from day 4, but there was no significant difference in the colon length of both groups (Fig. 1a, b).
Damage caused by ulceration and submucosal inflammation was identified by disruption of the epithelial barrier [27]. Histological analysis of the colons from untreated Dcir1 −/− mice showed intact epithelium, absence of edema, and normal muscle architecture (Fig. 1c). However, on day 8 after the induction of colitis, Dcir1 −/− mice showed milder colon tissue damage than WT mice, characterized by mucosal architecture, thickening of smooth muscle, presence of crypt abscesses, and cellular infiltration (Fig. 1c), particularly in the transverse and descending colon (Fig. 1c, the third and bottom row). In the cecum, increased mucosal erosion was observed in both groups, but inflammatory cell infiltration was greater in WT mice than in Dcir1 −/− mice (Fig. 1c, top row). The colitis scores of cecum and colon sections also supported the macroscopic observations (Fig. 1d). These data suggest that Dcir1 accelerates the development and/or promotes acute colitis.

Decreased infiltration of neutrophils during DSS colitis in Dcir1 −/− mice compared with WT mice

Massive infiltration of various types of leukocytes in the colon is often accompanied by the progression of DSS colitis [28]. In Dcir1 −/− mice, the infiltration of neutrophils into the colon was significantly lower than in WT mice (Fig. 2a), especially on day 5 (Fig. 2b). At the same time point, Ly6Chi, Ly6Cint, and Ly6Clo monocytic cells were slightly decreased in Dcir1 −/− mice compared with WT mice, although this was not statistically significant. Similarly, lower numbers of natural killer (NK) cells, NKT cells and T cells, but not B cells, infiltrated the colon in Dcir1 −/− mice compared with WT mice on day 5, but this was not statistically significant (Fig. 2c and d). Although both immuno-suppressive and -activating subtypes were present in the infiltrating cells, it is known that neutrophils are the first effector cells observed in the colon during the onset of DSS colitis. Collectively, these findings suggest that, in the Dcir1 expressing cells, a decrease of neutrophils rather than of monocytes, contributes to the amelioration of DSS colitis in Dcir1 −/− mice.

Decreased MIP-2 production in the intestine of Dcir1 −/− mice

We next checked cytokine/chemokine production in Dcir1 −/− and WT mouse intestines using a whole tissue culture method [29, 30]. Notably, on day 5 of DSS treatment, neutrophil chemoattractant MIP-2, not but KC, production in the colon and cecum of Dcir1 −/− mice was significantly lower than in WT mice, consistent with the reduced accumulation of neutrophils in the Dcir1 −/− colons (Figs. 2b, 3a). Although the production of monocyte chemoattractant MCP-1 and several proinflammatory cytokines, such as IFN-γ and IL-1β, tended to be lower in Dcir1 −/− colons, the difference was not statistically significant (Fig. 3a). The production of other cytokines was not different between the WT and Dcir1 −/− colons. We could not detect IL-12 expression. These data collectively suggest that Dcir1 may accelerate neutrophil recruitment through the enhanced MIP-2 production in inflamed colons.
To exclude the possibility that Dcir1 −/− neutrophils are less competent to respond to MIP-2, the chemotactic activity of bone marrow neutrophils was assessed using a transwell culture system. Results showed that a lack of Dcir1 did not affect the migration capacity of neutrophils in response to MIP-2 (Fig. 3b).

Decreased numbers of MPO+ neutrophils in the inflamed colon of Dcir1 −/− mice

MPO in neutrophils is involved in the production of ROS that are involved in both host defense and tissue injury [9, 10]. To identify the localization and numbers of MPO+ neutrophils, we stained cryosections of the colon using anti-MPO Ab in combination with anti-Ly6G and -CD11b mAbs on day 5. MPO+ neutrophils (CD11b+Ly6G+) infiltrated into the submucosal and mucosal areas of the colon in colitis-induced WT mice and Dcir1 −/− mice (Fig. 4a, arrowheads). However, fewer MPO+ neutrophils were accumulated in Dcir1 −/− mice colon than in WT mice colon (Fig. 4b). Without DSS treatment, there was no difference in the numbers of MPO+ neutrophils in WT and Dcir1 −/− colons. These results suggest that Dcir1 in WT mice enhances the infiltration of activated neutrophils into the colon, which exacerbates colitis.

Altered ROS production from Dcir1−/− neutrophils in response to LPS stimulation in vitro

We next focused on ROS production by neutrophils lacking Dcir1. Neutrophils were purified from bone marrow and stimulated with LPS (Fig. 5a). In comparison with WT neutrophils, Dcir1 −/− neutrophils produced lower amounts of ROS in response to LPS. However, Dcir1 −/− , but not WT, neutrophils produced more ROS in response to GM-CSF (Fig. 5b). This is consistent with a previous report that Dcir1 inhibited GM-CSF-dependent signaling [20]. These results suggest that Dcir1 enhances both infiltration of neutrophils into the inflamed colon, and their ability to produce ROS in response to microbes.

Discussion

Here we demonstrated that Dcir1 is involved in acute intestinal inflammation caused by DSS. Of note, both Dcir1 −/− and WT mice demonstrated similar body weight changes in the early days of DSS intake; however, Dcir1 −/− mice showed attenuated body weight loss from day 4 and a better clinical score especially in the transverse and distal colon compared with WT mice on day 8. These results imply that Dcir1 enhances the progression of DSS colitis.
MIP-2 production was lower in colons from Dcir1 −/− mice on day 5 of DSS colitis, when the number of neutrophils reached a peak. MIP-2 expression is correlated with the clinical activity index in human UC and murine DSS colitis [31, 32], and its receptor, CXCR2, is a major mediator of neutrophil influx in some disease models including acute colitis [33, 34]. The depletion or blocking of CXCR2 was protective against acute and chronic DSS colitis [35, 36]. Similarly, high levels of CXCR1/2 and CXCR1/2 ligand expression were observed in the mucosa of UC patients [37]. Although we observed the normal migration of bone marrow Dcir1 −/− neutrophils toward MIP-2, we found reduced accumulation of MPO+ neutrophils in colons on day 5 of DSS colitis. Activated neutrophils secrete MPO, resulting in mucosal disruption and ulceration [7, 38, 39]. Therefore, our data suggest that Dcir1 contributes to the development of intestinal inflammation though the recruitment of neutrophils and their MPO production. Furthermore, bone marrow neutrophils from Dcir1 −/− mice produced smaller amounts of ROS compared with WT neutrophils in response to LPS. During colitis, the epithelial barrier is damaged, leading to interactions between commensal bacteria and neutrophils. In this situation, Dcir1 −/− neutrophils may produce small amounts of ROS compared with WT neutrophils. Commensal bacteria have been implicated in the pathogenesis of human IBD and several animal models including DSS colitis [4042]. Therefore, a lack of Dcir1 might also contribute to ameliorating DSS colitis via the down modulation of ROS production.
We also found that a lack of Dcir1 enhanced GM-CSF signaling. Previous reports showed GM-CSF production in the colon of UC patients and DSS colitis-induced mice [43, 44]. The enhancement of GM-CSF signaling possibly suppresses colitis because GM-CSF −/− mice are more susceptible to DSS colitis [45] and GM-CSF treatment results in significantly improved clinical parameters and histology of DSS colitis [46]. These results suggest that a lack of Dcir1 enhances GM-CSF signaling, resulting in an improvement of DSS colitis. Taken together, a lack of Dcir1 causes reduced neutrophil accumulation via downregulation of MIP-2 expression, reduced ROS production and enhanced GM-CSF signaling, which possibly leads to an improvement of DSS induced colitis. In contrast, it is known that ROS production increases direct bactericidal activity [47] and neutrophil extracellular traps (NET) formation [48] by neutrophils. Lack of Dcir1 might affect these neutrophil functions.
We previously reported that Dcir1 −/− mice tended to develop autoimmune arthritis because of enhanced GM-CSF signaling and DCs development [20]. Furthermore, DCIR signaling induces the suppression of other pathways, such as B cell receptor signaling and proinflammatory cytokine production via TLR ligands [19, 49, 50]. However, Dcir1 enhances DSS colitis via several pathways as described here. Therefore, the balance between benefit and detriment caused by DCIR might be different in various types of disease models and experimental conditions. Recently, Hutter et al. reported no difference in DSS colitis symptoms observed between Dcir1 −/− and WT mice [51]. It was also reported that an increased cellular infiltration into the rectum of Dcir1 −/− mice was present at 7 days with 3 % DSS treatment. In our study, we used a different protocol (2.0 % DSS for 5 days followed by water for 3 days) for a more moderate induction of colitis, and observed an alleviation of body weight loss in Dcir1 −/− mice mainly after exchanging DSS for normal water. The simplest explanation for the different observations between studies is that the role of Dcir1 might be different depending on the symptoms of DSS colitis, or their use of stronger DSS colitis induction conditions compared with our study, which might have negated the benefit of a lack of Dcir1.

Conclusions

We showed that a lack of Dcir1 suppressed the development of experimental DSS colitis. In the intestine of Dcir1 −/− mice with colitis, the number of tissue-injuring MPO+ neutrophils was lower than in WT mice, possibly because of the down-regulation of MIP-2 chemokine. Furthermore, bone marrow neutrophils produced reduced levels of ROS in response to LPS. Our results suggest that Dcir1 affects the accumulation and properties of neutrophils, and enhances the development of DSS colitis. Further studies regarding the mechanisms controlling neutrophils by DCIR will provide new clues to understand intestinal inflammation.

Acknowledgements

We also acknowledge Dr. Takeshi Tanoue and Dr. Kenya Honda (RIKEN, IMS) for providing useful technical advice for LPMC isolation from mouse intestine.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

ST, KT, and KI designed the research, ST, MK and TI conducted the research and analyzed the data. ST and MK performed statistical analysis. ST, KI and KT wrote the paper and have primary responsibility for its all content. YI provided essential mouse strains, valuable advice and technical expertise for these studies. All authors read and approved the final manuscript.
Literatur
1.
Zurück zum Zitat Hanauer SB. Inflammatory bowel disease: epidemiology, pathogenesis, and therapeutic opportunities. Inflamm Bowel Dis. 2006;12 Suppl 1:S3–9.CrossRefPubMed Hanauer SB. Inflammatory bowel disease: epidemiology, pathogenesis, and therapeutic opportunities. Inflamm Bowel Dis. 2006;12 Suppl 1:S3–9.CrossRefPubMed
2.
3.
Zurück zum Zitat Kaser A, Zeissig S, Blumberg RS. Inflammatory bowel disease. Annu Rev Immunol. 2010;28:573–621.CrossRefPubMed Kaser A, Zeissig S, Blumberg RS. Inflammatory bowel disease. Annu Rev Immunol. 2010;28:573–621.CrossRefPubMed
4.
Zurück zum Zitat Maloy KJ, Powrie F. Intestinal homeostasis and its breakdown in inflammatory bowel disease. Nature. 2011;474:298–306.CrossRefPubMed Maloy KJ, Powrie F. Intestinal homeostasis and its breakdown in inflammatory bowel disease. Nature. 2011;474:298–306.CrossRefPubMed
5.
Zurück zum Zitat Cho JH, Brant SR. Recent insights into the genetics of inflammatory bowel disease. Gastroenterology. 2011;140:1704–12.CrossRefPubMed Cho JH, Brant SR. Recent insights into the genetics of inflammatory bowel disease. Gastroenterology. 2011;140:1704–12.CrossRefPubMed
6.
Zurück zum Zitat Abraham C, Medzhitov R. Interactions between the host innate immune system and microbes in inflammatory bowel disease. Gastroenterology. 2011;140:1729–37.PubMedCentralCrossRefPubMed Abraham C, Medzhitov R. Interactions between the host innate immune system and microbes in inflammatory bowel disease. Gastroenterology. 2011;140:1729–37.PubMedCentralCrossRefPubMed
7.
Zurück zum Zitat Fournier BM, Parkos CA. The role of neutrophils during intestinal inflammation. Mucosal Immunol. 2012;5:354–66.CrossRefPubMed Fournier BM, Parkos CA. The role of neutrophils during intestinal inflammation. Mucosal Immunol. 2012;5:354–66.CrossRefPubMed
8.
Zurück zum Zitat Hampton MB, Kettle AJ, Winterbourn CC. Inside the neutrophil phagosome: oxidants, myeloperoxidase, and bacterial killing. Blood. 1998;92:3007–17.PubMed Hampton MB, Kettle AJ, Winterbourn CC. Inside the neutrophil phagosome: oxidants, myeloperoxidase, and bacterial killing. Blood. 1998;92:3007–17.PubMed
9.
Zurück zum Zitat Elliott SN, Wallace JL. Neutrophil-mediated gastrointestinal injury. Can J Gastroenterol. 1998;12:559–68.PubMed Elliott SN, Wallace JL. Neutrophil-mediated gastrointestinal injury. Can J Gastroenterol. 1998;12:559–68.PubMed
10.
Zurück zum Zitat Hazen SL, Hsu FF, Mueller DM, Crowley JR, Heinecke JW. Human neutrophils employ chlorine gas as an oxidant during phagocytosis. J Clin Invest. 1996;98:1283–9.PubMedCentralCrossRefPubMed Hazen SL, Hsu FF, Mueller DM, Crowley JR, Heinecke JW. Human neutrophils employ chlorine gas as an oxidant during phagocytosis. J Clin Invest. 1996;98:1283–9.PubMedCentralCrossRefPubMed
11.
Zurück zum Zitat Takeuchi O, Akira S. Pattern recognition receptors and inflammation. Cell. 2010;140:805–20.CrossRefPubMed Takeuchi O, Akira S. Pattern recognition receptors and inflammation. Cell. 2010;140:805–20.CrossRefPubMed
12.
Zurück zum Zitat Walsh D, McCarthy J, O'Driscoll C, Melgar S. Pattern recognition receptors--molecular orchestrators of inflammation in inflammatory bowel disease. Cytokine Growth Factor Rev. 2013;24:91–104.CrossRefPubMed Walsh D, McCarthy J, O'Driscoll C, Melgar S. Pattern recognition receptors--molecular orchestrators of inflammation in inflammatory bowel disease. Cytokine Growth Factor Rev. 2013;24:91–104.CrossRefPubMed
13.
Zurück zum Zitat Bates EE, Fournier N, Garcia E, Valladeau J, Durand I, Pin JJ, et al. APCs express DCIR, a novel C-type lectin surface receptor containing an immunoreceptor tyrosine-based inhibitory motif. J Immunol. 1999;163:1973–83.PubMed Bates EE, Fournier N, Garcia E, Valladeau J, Durand I, Pin JJ, et al. APCs express DCIR, a novel C-type lectin surface receptor containing an immunoreceptor tyrosine-based inhibitory motif. J Immunol. 1999;163:1973–83.PubMed
14.
15.
Zurück zum Zitat Miyake Y, Ishikawa E, Ishikawa T, Yamasaki S. Self and nonself recognition through C-type lectin receptor. Mincle Self Nonself. 2010;1:310–3.CrossRefPubMed Miyake Y, Ishikawa E, Ishikawa T, Yamasaki S. Self and nonself recognition through C-type lectin receptor. Mincle Self Nonself. 2010;1:310–3.CrossRefPubMed
16.
Zurück zum Zitat Lambert AA, Gilbert C, Richard M, Beaulieu AD, Tremblay MJ. The C-type lectin surface receptor DCIR acts as a new attachment factor for HIV-1 in dendritic cells and contributes to trans- and cis-infection pathways. Blood. 2008;112:1299–307.PubMedCentralCrossRefPubMed Lambert AA, Gilbert C, Richard M, Beaulieu AD, Tremblay MJ. The C-type lectin surface receptor DCIR acts as a new attachment factor for HIV-1 in dendritic cells and contributes to trans- and cis-infection pathways. Blood. 2008;112:1299–307.PubMedCentralCrossRefPubMed
17.
Zurück zum Zitat Bloem K, Vuist IM, van den Berk M, Klaver EJ, van Die I, Knippels LM, et al. DCIR interacts with ligands from both endogenous and pathogenic origin. Immunol Lett. 2013;158:33–41.CrossRefPubMed Bloem K, Vuist IM, van den Berk M, Klaver EJ, van Die I, Knippels LM, et al. DCIR interacts with ligands from both endogenous and pathogenic origin. Immunol Lett. 2013;158:33–41.CrossRefPubMed
18.
Zurück zum Zitat Kaden SA, Kurig S, Vasters K, Hofmann K, Zaenker KS, Schmitz J, et al. Enhanced dendritic cell-induced immune responses mediated by the novel C-type lectin receptor mDCAR1. J Immunol. 2009;183:5069–78.CrossRefPubMed Kaden SA, Kurig S, Vasters K, Hofmann K, Zaenker KS, Schmitz J, et al. Enhanced dendritic cell-induced immune responses mediated by the novel C-type lectin receptor mDCAR1. J Immunol. 2009;183:5069–78.CrossRefPubMed
19.
Zurück zum Zitat Kanazawa N, Okazaki T, Nishimura H, Tashiro K, Inaba K, Miyachi Y. DCIR acts as an inhibitory receptor depending on its immunoreceptor tyrosine-based inhibitory motif. J Invest Dermatol. 2002;118:261–6.CrossRefPubMed Kanazawa N, Okazaki T, Nishimura H, Tashiro K, Inaba K, Miyachi Y. DCIR acts as an inhibitory receptor depending on its immunoreceptor tyrosine-based inhibitory motif. J Invest Dermatol. 2002;118:261–6.CrossRefPubMed
20.
Zurück zum Zitat Fujikado N, Saijo S, Yonezawa T, Shimamori K, Ishii A, Sugai S, et al. Dcir deficiency causes development of autoimmune diseases in mice due to excess expansion of dendritic cells. Nat Med. 2008;14:176–80.CrossRefPubMed Fujikado N, Saijo S, Yonezawa T, Shimamori K, Ishii A, Sugai S, et al. Dcir deficiency causes development of autoimmune diseases in mice due to excess expansion of dendritic cells. Nat Med. 2008;14:176–80.CrossRefPubMed
21.
Zurück zum Zitat Maglinao M, Klopfleisch R, Seeberger PH, Lepenies B. The C-type lectin receptor DCIR is crucial for the development of experimental cerebral malaria. J Immunol. 2013;191:2551–9.CrossRefPubMed Maglinao M, Klopfleisch R, Seeberger PH, Lepenies B. The C-type lectin receptor DCIR is crucial for the development of experimental cerebral malaria. J Immunol. 2013;191:2551–9.CrossRefPubMed
22.
Zurück zum Zitat Mitsuyama K, Toyonaga A, Sasaki E, Watanabe K, Tateishi H, Nishiyama T, et al. IL-8 as an important chemoattractant for neutrophils in ulcerative colitis and Crohn’s disease. Clin Exp Immunol. 1994;96:432–6.PubMedCentralCrossRefPubMed Mitsuyama K, Toyonaga A, Sasaki E, Watanabe K, Tateishi H, Nishiyama T, et al. IL-8 as an important chemoattractant for neutrophils in ulcerative colitis and Crohn’s disease. Clin Exp Immunol. 1994;96:432–6.PubMedCentralCrossRefPubMed
23.
Zurück zum Zitat Qualls JE, Kaplan AM, van Rooijen N, Cohen DA. Suppression of experimental colitis by intestinal mononuclear phagocytes. J Leukoc Biol. 2006;80:802–15.CrossRefPubMed Qualls JE, Kaplan AM, van Rooijen N, Cohen DA. Suppression of experimental colitis by intestinal mononuclear phagocytes. J Leukoc Biol. 2006;80:802–15.CrossRefPubMed
24.
Zurück zum Zitat Zigmond E, Varol C, Farache J, Elmaliah E, Satpathy AT, Friedlander G, et al. Ly6C hi monocytes in the inflamed colon give rise to proinflammatory effector cells and migratory antigen-presenting cells. Immunity. 2012;37:1076–90.CrossRefPubMed Zigmond E, Varol C, Farache J, Elmaliah E, Satpathy AT, Friedlander G, et al. Ly6C hi monocytes in the inflamed colon give rise to proinflammatory effector cells and migratory antigen-presenting cells. Immunity. 2012;37:1076–90.CrossRefPubMed
25.
Zurück zum Zitat Kojouharoff G, Hans W, Obermeier F, Mannel DN, Andus T, Scholmerich J, et al. Neutralization of tumour necrosis factor (TNF) but not of IL-1 reduces inflammation in chronic dextran sulphate sodium-induced colitis in mice. Clin Exp Immunol. 1997;107:353–8.PubMedCentralCrossRefPubMed Kojouharoff G, Hans W, Obermeier F, Mannel DN, Andus T, Scholmerich J, et al. Neutralization of tumour necrosis factor (TNF) but not of IL-1 reduces inflammation in chronic dextran sulphate sodium-induced colitis in mice. Clin Exp Immunol. 1997;107:353–8.PubMedCentralCrossRefPubMed
26.
Zurück zum Zitat Atarashi K, Tanoue T, Shima T, Imaoka A, Kuwahara T, Momose Y, et al. Induction of colonic regulatory T cells by indigenous Clostridium species. Science. 2011;331:337–41.PubMedCentralCrossRefPubMed Atarashi K, Tanoue T, Shima T, Imaoka A, Kuwahara T, Momose Y, et al. Induction of colonic regulatory T cells by indigenous Clostridium species. Science. 2011;331:337–41.PubMedCentralCrossRefPubMed
27.
Zurück zum Zitat Okayasu I, Hatakeyama S, Yamada M, Ohkusa T, Inagaki Y, Nakaya R. A novel method in the induction of reliable experimental acute and chronic ulcerative colitis in mice. Gastroenterology. 1990;98:694–702.PubMed Okayasu I, Hatakeyama S, Yamada M, Ohkusa T, Inagaki Y, Nakaya R. A novel method in the induction of reliable experimental acute and chronic ulcerative colitis in mice. Gastroenterology. 1990;98:694–702.PubMed
29.
Zurück zum Zitat Sellon RK, Tonkonogy S, Schultz M, Dieleman LA, Grenther W, Balish E, et al. Resident enteric bacteria are necessary for development of spontaneous colitis and immune system activation in interleukin-10-deficient mice. Infect Immun. 1998;66:5224–31.PubMedCentralPubMed Sellon RK, Tonkonogy S, Schultz M, Dieleman LA, Grenther W, Balish E, et al. Resident enteric bacteria are necessary for development of spontaneous colitis and immune system activation in interleukin-10-deficient mice. Infect Immun. 1998;66:5224–31.PubMedCentralPubMed
30.
Zurück zum Zitat Clarke CJ, Stokes CR. The intestinal and serum humoral immune response of mice to orally administered antigens in liposomes: II. The response to liposome-entrapped bacterial proteins. Vet Immunol Immunopathol. 1992;32:139–48.CrossRefPubMed Clarke CJ, Stokes CR. The intestinal and serum humoral immune response of mice to orally administered antigens in liposomes: II. The response to liposome-entrapped bacterial proteins. Vet Immunol Immunopathol. 1992;32:139–48.CrossRefPubMed
31.
Zurück zum Zitat Ohtsuka Y, Sanderson IR. Dextran sulfate sodium-induced inflammation is enhanced by intestinal epithelial cell chemokine expression in mice. Pediatr Res. 2003;53:143–7.PubMed Ohtsuka Y, Sanderson IR. Dextran sulfate sodium-induced inflammation is enhanced by intestinal epithelial cell chemokine expression in mice. Pediatr Res. 2003;53:143–7.PubMed
32.
Zurück zum Zitat Zahn A, Giese T, Karner M, Braun A, Hinz U, Stremmel W, et al. Transcript levels of different cytokines and chemokines correlate with clinical and endoscopic activity in ulcerative colitis. BMC Gastroenterol. 2009;9:13.PubMedCentralCrossRefPubMed Zahn A, Giese T, Karner M, Braun A, Hinz U, Stremmel W, et al. Transcript levels of different cytokines and chemokines correlate with clinical and endoscopic activity in ulcerative colitis. BMC Gastroenterol. 2009;9:13.PubMedCentralCrossRefPubMed
33.
Zurück zum Zitat Podolin PL, Bolognese BJ, Foley JJ, Schmidt DB, Buckley PT, Widdowson KL, et al. A potent and selective nonpeptide antagonist of CXCR2 inhibits acute and chronic models of arthritis in the rabbit. J Immunol. 2002;169:6435–44.CrossRefPubMed Podolin PL, Bolognese BJ, Foley JJ, Schmidt DB, Buckley PT, Widdowson KL, et al. A potent and selective nonpeptide antagonist of CXCR2 inhibits acute and chronic models of arthritis in the rabbit. J Immunol. 2002;169:6435–44.CrossRefPubMed
34.
Zurück zum Zitat Ajuebor MN, Zagorski J, Kunkel SL, Strieter RM, Hogaboam CM. Contrasting roles for CXCR2 during experimental colitis. Exp Mol Pathol. 2004;76:1–8.CrossRefPubMed Ajuebor MN, Zagorski J, Kunkel SL, Strieter RM, Hogaboam CM. Contrasting roles for CXCR2 during experimental colitis. Exp Mol Pathol. 2004;76:1–8.CrossRefPubMed
35.
Zurück zum Zitat Buanne P, Di Carlo E, Caputi L, Brandolini L, Mosca M, Cattani F, et al. Crucial pathophysiological role of CXCR2 in experimental ulcerative colitis in mice. J Leukoc Biol. 2007;82:1239–46.CrossRefPubMed Buanne P, Di Carlo E, Caputi L, Brandolini L, Mosca M, Cattani F, et al. Crucial pathophysiological role of CXCR2 in experimental ulcerative colitis in mice. J Leukoc Biol. 2007;82:1239–46.CrossRefPubMed
36.
Zurück zum Zitat Farooq SM, Stillie R, Svensson M, Svanborg C, Strieter RM, Stadnyk AW. Therapeutic effect of blocking CXCR2 on neutrophil recruitment and dextran sodium sulfate-induced colitis. J Pharmacol Exp Ther. 2009;329:123–9.CrossRefPubMed Farooq SM, Stillie R, Svensson M, Svanborg C, Strieter RM, Stadnyk AW. Therapeutic effect of blocking CXCR2 on neutrophil recruitment and dextran sodium sulfate-induced colitis. J Pharmacol Exp Ther. 2009;329:123–9.CrossRefPubMed
37.
Zurück zum Zitat Williams EJ, Haque S, Banks C, Johnson P, Sarsfield P, Sheron N. Distribution of the interleukin-8 receptors, CXCR1 and CXCR2, in inflamed gut tissue. J Pathol. 2000;192:533–9.CrossRefPubMed Williams EJ, Haque S, Banks C, Johnson P, Sarsfield P, Sheron N. Distribution of the interleukin-8 receptors, CXCR1 and CXCR2, in inflamed gut tissue. J Pathol. 2000;192:533–9.CrossRefPubMed
38.
Zurück zum Zitat McKenzie SJ, Baker MS, Buffinton GD, Doe WF. Evidence of oxidant-induced injury to epithelial cells during inflammatory bowel disease. J Clin Invest. 1996;98:136–41.PubMedCentralCrossRefPubMed McKenzie SJ, Baker MS, Buffinton GD, Doe WF. Evidence of oxidant-induced injury to epithelial cells during inflammatory bowel disease. J Clin Invest. 1996;98:136–41.PubMedCentralCrossRefPubMed
39.
Zurück zum Zitat Fiocchi C. Inflammatory bowel disease: etiology and pathogenesis. Gastroenterology. 1998;115:182–205.CrossRefPubMed Fiocchi C. Inflammatory bowel disease: etiology and pathogenesis. Gastroenterology. 1998;115:182–205.CrossRefPubMed
41.
Zurück zum Zitat Kim SC, Tonkonogy SL, Albright CA, Tsang J, Balish EJ, Braun J, et al. Variable phenotypes of enterocolitis in interleukin 10-deficient mice monoassociated with two different commensal bacteria. Gastroenterology. 2005;128:891–906.CrossRefPubMed Kim SC, Tonkonogy SL, Albright CA, Tsang J, Balish EJ, Braun J, et al. Variable phenotypes of enterocolitis in interleukin 10-deficient mice monoassociated with two different commensal bacteria. Gastroenterology. 2005;128:891–906.CrossRefPubMed
42.
Zurück zum Zitat Nakanishi Y, Sato T, Ohteki T. Commensal Gram-positive bacteria initiates colitis by inducing monocyte/macrophage mobilization. Mucosal Immunol. 2015;8:152–60.CrossRefPubMed Nakanishi Y, Sato T, Ohteki T. Commensal Gram-positive bacteria initiates colitis by inducing monocyte/macrophage mobilization. Mucosal Immunol. 2015;8:152–60.CrossRefPubMed
43.
Zurück zum Zitat Noguchi M, Hiwatashi N, Liu ZX, Toyota T. Increased secretion of granulocyte-macrophage colony-stimulating factor in mucosal lesions of inflammatory bowel disease. Digestion. 2001;63 Suppl 1:32–6.CrossRefPubMed Noguchi M, Hiwatashi N, Liu ZX, Toyota T. Increased secretion of granulocyte-macrophage colony-stimulating factor in mucosal lesions of inflammatory bowel disease. Digestion. 2001;63 Suppl 1:32–6.CrossRefPubMed
44.
Zurück zum Zitat Egea L, McAllister CS, Lakhdari O, Minev I, Shenouda S, Kagnoff MF. GM-CSF produced by nonhematopoietic cells is required for early epithelial cell proliferation and repair of injured colonic mucosa. J Immunol. 2013;190:1702–13.PubMedCentralCrossRefPubMed Egea L, McAllister CS, Lakhdari O, Minev I, Shenouda S, Kagnoff MF. GM-CSF produced by nonhematopoietic cells is required for early epithelial cell proliferation and repair of injured colonic mucosa. J Immunol. 2013;190:1702–13.PubMedCentralCrossRefPubMed
45.
Zurück zum Zitat Xu Y, Hunt NH, Bao S. The role of granulocyte macrophage-colony-stimulating factor in acute intestinal inflammation. Cell Res. 2008;18:1220–9.CrossRefPubMed Xu Y, Hunt NH, Bao S. The role of granulocyte macrophage-colony-stimulating factor in acute intestinal inflammation. Cell Res. 2008;18:1220–9.CrossRefPubMed
46.
Zurück zum Zitat Sainathan SK, Hanna EM, Gong Q, Bishnupuri KS, Luo Q, Colonna M, et al. Granulocyte macrophage colony-stimulating factor ameliorates DSS-induced experimental colitis. Inflamm Bowel Dis. 2008;14:88–99.PubMedCentralCrossRefPubMed Sainathan SK, Hanna EM, Gong Q, Bishnupuri KS, Luo Q, Colonna M, et al. Granulocyte macrophage colony-stimulating factor ameliorates DSS-induced experimental colitis. Inflamm Bowel Dis. 2008;14:88–99.PubMedCentralCrossRefPubMed
48.
Zurück zum Zitat Metzler KD, Fuchs TA, Nauseef WM, Reumaux D, Roesler J, Schulze I, et al. Myeloperoxidase is required for neutrophil extracellular trap formation: implications for innate immunity. Blood. 2011;117:953–9.PubMedCentralCrossRefPubMed Metzler KD, Fuchs TA, Nauseef WM, Reumaux D, Roesler J, Schulze I, et al. Myeloperoxidase is required for neutrophil extracellular trap formation: implications for innate immunity. Blood. 2011;117:953–9.PubMedCentralCrossRefPubMed
49.
Zurück zum Zitat Meyer-Wentrup F, Benitez-Ribas D, Tacken PJ, Punt CJ, Figdor CG, de Vries IJ, et al. Targeting DCIR on human plasmacytoid dendritic cells results in antigen presentation and inhibits IFN-α production. Blood. 2008;111:4245–53.CrossRefPubMed Meyer-Wentrup F, Benitez-Ribas D, Tacken PJ, Punt CJ, Figdor CG, de Vries IJ, et al. Targeting DCIR on human plasmacytoid dendritic cells results in antigen presentation and inhibits IFN-α production. Blood. 2008;111:4245–53.CrossRefPubMed
50.
Zurück zum Zitat Meyer-Wentrup F, Cambi A, Joosten B, Looman MW, de Vries IJ, Figdor CG, et al. DCIR is endocytosed into human dendritic cells and inhibits TLR8-mediated cytokine production. J Leukoc Biol. 2009;85:518–25.CrossRefPubMed Meyer-Wentrup F, Cambi A, Joosten B, Looman MW, de Vries IJ, Figdor CG, et al. DCIR is endocytosed into human dendritic cells and inhibits TLR8-mediated cytokine production. J Leukoc Biol. 2009;85:518–25.CrossRefPubMed
51.
Zurück zum Zitat Hutter J, Eriksson M, Johannssen T, Klopfleisch R, von Smolinski D, Gruber AD, et al. Role of the C-type lectin receptors MCL and DCIR in experimental colitis. PLoS One. 2014;9:e103281.PubMedCentralCrossRefPubMed Hutter J, Eriksson M, Johannssen T, Klopfleisch R, von Smolinski D, Gruber AD, et al. Role of the C-type lectin receptors MCL and DCIR in experimental colitis. PLoS One. 2014;9:e103281.PubMedCentralCrossRefPubMed
Metadaten
Titel
Dendritic cell immunoreceptor 1 alters neutrophil responses in the development of experimental colitis
verfasst von
Sumika Tokieda
Marie Komori
Toshifumi Ishiguro
Yoichiro Iwakura
Kazuhiko Takahara
Kayo Inaba
Publikationsdatum
01.12.2015
Verlag
BioMed Central
Erschienen in
BMC Immunology / Ausgabe 1/2015
Elektronische ISSN: 1471-2172
DOI
https://doi.org/10.1186/s12865-015-0129-5

Weitere Artikel der Ausgabe 1/2015

BMC Immunology 1/2015 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.