Characterization of the effect of LPS on dendritic cell subset discrimination in spleen

Similar documents
Supporting Information

Supplementary Figure 1. Efficient DC depletion in CD11c.DOG transgenic mice

Primary Adult Naïve CD4+ CD45RA+ Cells. Prepared by: David Randolph at University of Alabama, Birmingham

Blocking antibodies and peptides. Rat anti-mouse PD-1 (29F.1A12, rat IgG2a, k), PD-

Nature Immunology: doi: /ni Supplementary Figure 1. Examples of staining for each antibody used for the mass cytometry analysis.

In vitro human regulatory T cell expansion

SUPPORTING INFORMATIONS

Antigen presenting capacity of murine splenic myeloid cells

MagniSort Mouse CD4 Naive T cell Enrichment Kit Catalog Number: RUO: For Research Use Only. Not for use in diagnostic procedures.

Identification of a novel antigen cross-presenting cell type in spleen

In vitro human regulatory T cell expansion

NK cell flow cytometric assay In vivo DC viability and migration assay

Direct ex vivo characterization of human antigen-specific CD154 + CD4 + T cells Rapid antigen-reactive T cell enrichment (Rapid ARTE)

Rapid antigen-specific T cell enrichment (Rapid ARTE)

Supporting Online Material for

MATERIALS AND METHODS. Neutralizing antibodies specific to mouse Dll1, Dll4, J1 and J2 were prepared as described. 1,2 All

SUPPLEMENTARY INFORMATION

In vitro human regulatory T cell suppression assay

SUPPLEMENTARY INFORMATION

BMDCs were generated in vitro from bone marrow cells cultured in 10 % RPMI supplemented

MagniSort Mouse CD4 T cell Enrichment Kit Catalog Number: RUO: For Research Use Only. Not for use in diagnostic procedures.

T H 1, T H 2 and T H 17 polarization of naïve CD4 + mouse T cells

Suppl Video: Tumor cells (green) and monocytes (white) are seeded on a confluent endothelial

COMPONENT NAME COMPONENT # QUANTITY STORAGE SHELF LIFE FORMAT. Store at 2-8 C. Do not freeze. Store at 2-8 C. Do not freeze.

Increased IL-12 induced STAT-4 signaling in CD8 T cells. from aged mice

Combined Rho-kinase inhibition and immunogenic cell death triggers and propagates immunity against cancer

L-selectin Is Essential for Delivery of Activated CD8 + T Cells to Virus-Infected Organs for Protective Immunity

Detailed step-by-step operating procedures for NK cell and CTL degranulation assays

90 min 18 min. 45 min. 14 d

Natural Killer T Cells Are Critical for Dendritic Cells to Induce Immunity in Chlamydial Pneumonia

COMPONENT NAME COMPONENT # QUANTITY STORAGE SHELF LIFE FORMAT. Store at 2-8 C. Do not freeze. Store at 2-8 C. Do not freeze.

COMPONENT NAME COMPONENT # QUANTITY STORAGE SHELF LIFE FORMAT. Store at 2-8 C. Do not freeze. Store at 2-8 C. Do not freeze.

Stroma-dependent development of two dendritic-like cell types with distinct antigen presenting capability

Question 1. Kupffer cells, microglial cells and osteoclasts are all examples of what type of immune system cell?

SUPPLEMENTARY METHODS

Institute t of Experimental Immunology University of Bonn, Germany

Comprehensive evaluation of human immune system reconstitution in NSG. and NSG -SGM3 mouse models toward the development of a novel ONCO-HU

Hua Tang, Weiping Cao, Sudhir Pai Kasturi, Rajesh Ravindran, Helder I Nakaya, Kousik

BD IMag Cell Separation System

MCSF drives regulatory DC development in stromal co-cultures supporting hematopoiesis

TCR, MHC and coreceptors

W/T Itgam -/- F4/80 CD115. F4/80 hi CD115 + F4/80 + CD115 +

MagniSort Human CD4 Memory T cell Enrichment Kit Catalog Number: RUO: For Research Use Only. Not for use in diagnostic procedures.

Cell isolation. Spleen and lymph nodes (axillary, inguinal) were removed from mice

Dendritic cells and their role in immunity

Supplementary Figure 1. Characterization of basophils after reconstitution of SCID mice

Differential Response of Respiratory Dendritic Cell Subsets to Influenza Virus Infection

Technical Advance: In vitro production of distinct dendritic-like antigen-presenting cells from self-renewing hematopoietic stem cells

Efficient Isolation of Mouse Liver NKT Cells by Perfusion

Hematopoiesis. Hematopoiesis. Hematopoiesis

FOR OPTIMAL GUT HEALTH KEMIN.COM/GUTHEALTH

Application Information Bulletin: Human NK Cells Phenotypic characterizing of human Natural Killer (NK) cell populations in peripheral blood

Supplementary Table; Supplementary Figures and legends S1-S21; Supplementary Materials and Methods

VEGFR2-Mediated Vascular Dilation as a Mechanism of VEGF-Induced Anemia and Bone Marrow Cell Mobilization

Supplementary Figure S1. Flow cytometric analysis of the expression of Thy1 in NH cells. Flow cytometric analysis of the expression of T1/ST2 and

MACROPHAGE "MONOCYTES" SURFACE RECEPTORS

T-cell activation T cells migrate to secondary lymphoid tissues where they interact with antigen, antigen-presenting cells, and other lymphocytes:

T-cell activation T cells migrate to secondary lymphoid tissues where they interact with antigen, antigen-presenting cells, and other lymphocytes:

Antigen Presentation and T Lymphocyte Activation. Abul K. Abbas UCSF. FOCiS

Serafino et al. Thymosin α1 activates complement receptor-mediated phagocytosis in human monocyte-derived macrophages. SUPPLEMENTARY FIGURES

Eosinophils are required. for the maintenance of plasma cells in the bone marrow

PBS Class #2 Introduction to the Immune System part II Suggested reading: Abbas, pgs , 27-30

B6/COLODR/SPL/11C/83/LAP/#2.006 B6/COLODR/SPL/11C/86/LAP/#2.016 CD11C B6/COLODR/SPL/11C/80/LAP/#2.011 CD11C

MagniSort Human CD4 T cell Enrichment Kit Catalog Number: RUO: For Research Use Only. Not for use in diagnostic procedures.

Supplementary Materials for

Page 1 of 2. Product Information Contents: ezkine Th1 Activation 2 Whole Blood Intracellular Cytokine Kit

M.Sc. III Semester Biotechnology End Semester Examination, 2013 Model Answer LBTM: 302 Advanced Immunology

Supporting Information

Concentration Estimation from Flow Cytometry Exosome Data Protocol

Advances in Cancer Immunotherapy

IL-6Rα IL-6RαT-KO KO. IL-6Rα f/f bp. f/f 628 bp deleted 368 bp. 500 bp

Figure S1. Generation of inducible PTEN deficient mice and the BMMCs (A) B6.129 Pten loxp/loxp mice were mated with B6.

Figure S1. Western blot analysis of clathrin RNA interference in human DCs Human immature DCs were transfected with 100 nm Clathrin SMARTpool or

Supplemental Information. Aryl Hydrocarbon Receptor Controls. Monocyte Differentiation. into Dendritic Cells versus Macrophages

Cover Page. The handle holds various files of this Leiden University dissertation.

Ex vivo Human Antigen-specific T Cell Proliferation and Degranulation Willemijn Hobo 1, Wieger Norde 1 and Harry Dolstra 2*

Interferon γ regulates idiopathic pneumonia syndrome, a. Th17 + CD4 + T-cell-mediated GvH disease

a surface permeabilized

1. Overview of Adaptive Immunity

Supplemental Table I.

ANATOMY OF THE IMMUNE SYSTEM

Supplementary Figures

Measuring Dendritic Cells

Supplementary Figure 1. Immune profiles of untreated and PD-1 blockade resistant EGFR and Kras mouse lung tumors (a) Total lung weight of untreated

What is the immune system? Types of Immunity. Pasteur and rabies vaccine. Historical Role of smallpox. Recognition Response

Supporting Information

Figure S1. PMVs from THP-1 cells expose phosphatidylserine and carry actin. A) Flow

BD Flow Cytometry Reagents Multicolor Panels Designed for Optimal Resolution with the BD LSRFortessa X-20 Cell Analyzer

Instructions for Use. APO-AB Annexin V-Biotin Apoptosis Detection Kit 100 tests

Overview of the Lymphoid System

Evaluation of directed and random motility in microslides Assessment of leukocyte adhesion in flow chambers

Horizon 2020 Programme. SFS-01b Tackling losses from terrestrial animal diseases

sequences of a styx mutant reveals a T to A transversion in the donor splice site of intron 5

ezkine Th1/Th17 Whole Blood Intracellular Cytokine Kit Catalog Number: RUO: For Research Use Only. Not for use in diagnostic procedures.

Phagocytosis of FITC labelled opsonized and non-opsonized E. coli bacteria by monocytes and granulocytes in a whole blood assay

Fluorochrome Panel 1 Panel 2 Panel 3 Panel 4 Panel 5 CTLA-4 CTLA-4 CD15 CD3 FITC. Bio) PD-1 (MIH4, BD) ICOS (C398.4A, Biolegend) PD-L1 (MIH1, BD)

IMMUNOLOGY. Source, Isolate, Culture, And Analyze Immune Cells. Scientists Helping Scientists

Online supplement. Phenotypic, functional and plasticity features of classical and alternatively activated

Frontiers of Science Foundation Scholar. Toby Bothwell. Freshman, University of Arkansas. final research manuscripts

Transcription:

Short Communication J. Cell. Mol. Med. Vol 18, No 9, 2014 pp. 1908-1912 Characterization of the effect of LPS on dendritic cell subset discrimination in spleen Kristin L Griffiths, Jonathan KH Tan, Helen C O Neill * Abstract Research School of Biology, The Australian National University, Canberra, ACT, Australia Received: November 6, 2013; Accepted: May 7, 2014 The Gram-negative bacterial endotoxin lipopolysaccharide (LPS) is a potent inflammatory mediator and a leading cause of bacterial sepsis. While LPS is known to activate antigen-presenting cells, here we find that LPS down-regulates expression of CD11c and CD11b on splenic dendritic cell subsets, thus confounding the ability to identify these subsets following treatment. This has implications with regard to tracking the response to LPS in terms of the cell subsets involved, and should be considered whenever such studies are undertaken. Keywords: dendritic cell lipopolysaccharide spleen Introduction Dendritic cells (DCs) are often considered the sentinels of the immune system, acting as antigen-presenting cells (APC) with the primary role of taking up antigen and presenting it to T cells to initiate an adaptive immune response. It is now accepted that DCs can be divided into several different subsets based on marker expression, function and tissue/organ of residence (reviewed by Merad et al. [1]). Two of the key markers used to identify dendritic and myeloid cells from other leucocytes are CD11b and CD11c. As b 2 integrins, both exist as a heterodimer with CD18 on the cell surface [2], and both are thought to have roles in adhesion to endothelial cells and in complement recognition. CD11b, in particular, has been implicated in leucocyte rolling and crawling, mechanisms employed to capture and slow leucocytes flowing through blood vessels to facilitate extravasation [3]. In relation to DC identification, CD11c and CD11b are used as two of the primary markers for delineation of splenic DC subsets [1]. The Gram-negative bacterial endotoxin lipopolysaccharide (LPS) is known to be a potent inflammatory mediator. Here, we use LPS as a model toxin to induce inflammation in vivo to investigate the effects of the toxin on marker expression and prevalence of APC subsets in the spleen. Ability to identify APC subsets throughout the lymphoid system is important for the dissection of the immune response to *Correspondence to: Professor Helen C O NEILL, Research School of Biology, Bldg 134 Linnaeus Way, The Australian National University, Canberra, ACT 2600, Australia. Tel.: +612 6125 4720 Fax: +612 6125 0313 E-mail: helen.oneill@anu.edu.au infection. This paper identifies shortcomings of performing analyses of APC subset representation under inflammatory conditions. Materials and methods Animals Specific-pathogen free 6-week-old female C57BL/6J mice were obtained from the John Curtin School of Medical Research (Australian National University, Canberra, Australia). Mice were housed and handled according to protocols approved by the Animal Experimentation Ethics Committee at the Australian National University and killed by cervical dislocation. LPS treatment of mice Mice were treated through i.v. injection of LPS in HBSS at a dose of 0.1 lg/g bodyweight. Injections were performed into the tail vein of mice placed in a restraint by using a 1-ml syringe equipped with a 26G needle. Preparation of lymphoid cells For preparation of single-cell suspensions, dissected spleen or mesenteric lymph nodes were pressed through a fine mesh sieve. Cells were resuspended in 5 ml DMEM supplemented with 10% Foetal Calf Serum (JRH Biosciences, Lenexa, KS, USA), 10 mm Hepes (JRH Biosciences), 2 mm L-glutamine (JRH Biosciences), 100 lg penicillin/streptomycin doi: 10.1111/jcmm.12332 This is an open access article under the terms of the Creative Commons Attribution License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited.

J. Cell. Mol. Med. Vol 18, No 9, 2014 and 5 9 10 5 M 2-mercaptoethanol (BDH Ltd., Poole, UK; sdmem). Cells were then sedimented, and splenocytes were treated with RBC lysis buffer (140 mm NH 4 Cl, 17 mm Tris base) and washed. MACS separation Enrichment of DC populations was performed by depletion of T and B cells from a cell suspension of dissociated spleen. Cells were incubated with 0.25 lg/ml biotinylated anti-cd19 (B cells: ebioscience; San Diego, CA, USA) and 0.2 lg/ml biotinylated anti-thy1.2 (T cells: ebioscience) antibody per 10 8 cells in 1 ml. Antibody was diluted in MACS labelling buffer (PBS/2 mm EDTA/5% BSA) and absorbed to cells for 10 min. on ice Cells were washed twice with labelling buffer before addition of antibiotin magnetic microbeads (Miltenyi Biotec, Auburn, CA, USA). Cells were absorbed with 13 ll beads/10 8 cells in 1 ml for 25 min. on ice. After washing with MACS labelling buffer, cells were resuspended in 500 ll of buffer. The cell suspension was then run through a prewashed LS column (Miltenyi Biotec) positioned in the strong magnetic field of a SuperMACS II Separator (Miltenyi Biotec), which retains cells with bound magnetic microbeads. Splenocytes depleted of T and B cells were collected as flow-through cells, along with cells collected after three column washes with 3 ml labelling buffer. These cells were resuspended in sdmem for staining. Antibody staining of cells Cells were sedimented into the wells of a flexible 96-well polystyrene microtitre plate (Corning, New York, USA). For FcR blocking, cells were resuspended in 25 ll anti-cd16/32 followed by incubation on ice for 15 min. After washing, cells were resuspended in a primary antibody cocktail (APC-conjugated CD11c (N418), PE-Cy7-conjugated CD11b (M1/ 70), PE-conjugated CD8a (53-6.7), biotin- or FITC-conjugated MHC-II (AF6-120.1) all from ebioscience) and incubated on ice for 25 min. before washing. Where necessary, secondary labelled conjugate (streptavidin-apc-cy7 from ebioscience) was added and cells were incubated for 25 min. on ice, washed and transferred to a cluster tube (Corning) for analysis performed with a flow cytometer (Becton Dickinson LSRII, San Jose, CA, USA). Prior to analysis, propidium iodide (PI: 1000 lg/ ml) was added for dead cell discrimination. Flow cytometry Flow cytometry was performed with an LSRII flow cytometer (Becton Dickinson). Background binding of each antibody was determined by using isotype control antibodies corresponding to each fluorescenceconjugated antibody used, and gates were set by using this information as well as internal positive and negative cell subsets. Live cells were detected by absence of staining with PI. Analysis was performed with BD FACSDiva Software (Becton Dickinson) and Flow Jo software (Tristar, Phoenix, AZ, USA). Endocytic capacity of cells Ovalbumin (OVA)-FITC (100 lg/g bodyweight) was injected i.v. into mice in HBSS. Mice were left for 24 hrs before killing and spleen removal. Spleens were dissociated and stained with antibodies as above. Endocytosis was measured as% FITC + cells. Results LPS induces down-regulation of CD11c and CD11b expression on DC populations in spleen In this paper, we take steady-state splenic dendritic and myeloid cell subsets to include CD8a + and CD8a conventional DCs (cdcs), plasmacytoid-pre-dcs (p-predcs), monocytes and L-DCs (described by Tan et al. in 2011 [4]). Figure 1A shows clear identification of splenic dendritic and myeloid subsets on a CD11c versus CD11b plot in untreated C57BL/6J mice, confirming that in the steady-state, each splenic dendritic/myeloid subset has unique CD11c and CD11b expression levels. We next investigated the effect of endotoxin treatment on surface expression of CD11c, CD11b, CD8a and MHC-II on splenic dendritic and myeloid cell subsets. C57BL/6J mice were administered LPS i.v. and subsets analysed as above. As shown in Figure 1B, the greatest effect of LPS treatment on cdc subsets was to reduce levels of both CD11c and CD11b expression. This resulted in the cdc subsets being indistinguishable on the basis of CD11c and CD11b expression. L- DCs and monocytes have low or no CD11c expression, respectively, and this remains the case following LPS treatment except for a slight increase in CD11c expression on a subset of L-DC. Similarly, these subsets maintain high, but slightly lower, levels of CD11b expression. LPS treatment up-regulated MHC-II on the cell surface of all subsets except CD8 cdcs in terms of expression of markers investigated here (Fig 1B and Table 1). LPS decreases relative percentage of p-predcs in spleen To determine the effect of endotoxin treatment on the relative prevalence of splenic dendritic and myeloid cell subsets, representation of each subset as a percentage of the total dendritic and myeloid population in spleen was calculated (Fig. 2A). Interestingly, none of the mature subsets showed any change in prevalence following LPS treatment. Relative percentages of p-predcs, on the other hand, were reduced under conditions of simulated inflammation (Fig. 2A). To assess possible p-predc migration to lymph nodes as mature pdcs, percentages of CD8a + cdcs, CD8a cdcs and pdcs in mesenteric lymph nodes were determined. Results showed an increased percentages of CD8a cdcs and a small, but non-significant, decrease in pdcs (Fig. 2B). The effect of LPS on in vivo endocytic capacity of cells We next investigated the effect of LPS treatment on the endocytic capacity of dendritic and myeloid cells in the spleen. Mice were admin- 1909

A B 1910

J. Cell. Mol. Med. Vol 18, No 9, 2014 Fig. 1 Gating protocol used to assess the effect of LPS on marker expression on myeloid and dendritic cell subsets in spleen. (A) Spleens from 6-week-old C57BL/6J mice were depleted of T and B cells by using magnetic bead separation and stained by using fluorochrome-conjugated antibodies for analysis by flow cytometry. After dead cell exclusion based on propidium iodide (PI) staining, dendritic and myeloid cell subsets were identified by gating on CD11c versus CD8 and then CD11b versus MHC-II plots as described in Tan et al. [4]. Each identified population was then overlaid on a CD11b versus CD11c plot. (B) Six-week-old C57BL/6J mice were injected i.v. with LPS (0.1 lg/g bodyweight) 24 hrs prior to killing. Splenocytes were prepared and gating performed as described above. Changes in MHC-II expression on subsets is revealed by overlaid histograms: treated (coloured line); untreated (grey filled). Table 1 MHC-II expression increases following LPS treatment DC subset MHC-II (MFI: mean SD) Control (n = 7) LPS (n = 5) CD8a+ cdcs 59.7 16.0 231.0 69.6*** CD8a- cdcs 80.0 21.8 122.1 45.4 p-predcs 3.7 0.3 9.73 1.8*** Monocytes 2.8 0.4 6.7 2.2** L-DCs 3.1 2.4 40.1 14.8*** MFI: mean fluorescence intensity. **P < 0.01, ***P < 0.001. istered LPS i.v. with or without FITC-conjugated ovalbumin (OVA-FITC). Spleens were taken 24 hrs later and the amount of OVA-FITC taken up by each cell subset examined by flow cytometry. Results (Fig. 2C) showed that, with the exception of monocytes and CD8a + cdcs, LPS had no effect on the in vivo endocytic capacity of cells. Monocytes showed some increase in uptake of OVA-FITC, while CD8a + cdcs had slightly reduced endocytic capacity following LPS treatment. The increase in phagocytic capacity of myeloid cells could reflect a form of maturation towards a macrophage induced by LPS, whose primary function is phagocytosis for destruction of an invading pathogen. Indeed, functionally, LPS has been shown to induce phagocytosis in macrophages [5, 6], an observation supported by data presented here. It is likely, however, that CD8a + cdcs, on the other hand, respond to maturation by reducing levels of phagocytosis so as to divert energy into antigen presentation following encounter with the pathogen. A B C Fig. 2 The effect of LPS on prevalence and endocytic capacity of dendritic and myeloid subsets. (A) Splenocytes from LPS-treated (closed symbols) and untreated (open symbols) animals were prepared, stained and analysed by flow cytometry as described for Figure 1. Percentage representation of each dendritic and myeloid subset relative to total dendritic and myeloid cells was calculated and depicted graphically. (B) Mesenteric lymph nodes from LPS-treated and untreated animals were prepared, stained and gated as described for spleens, with the exception that no T- and B-cell depletion was performed. Relative percentage of each dendritic and myeloid subset was calculated and graphed. (C) C57BL/6J mice were given OVA-FITC (100 lg/g bodyweight) along with LPS (0.1 lg/g bodyweight) at 24 hrs prior to killing. Splenocytes were prepared as described in Figure 1, and following identification of each dendritic and myeloid subset, uptake of OVA-FITC was measured. Dotted lines represent OVA-FITC uptake by cells from untreated mice; solid lines represent OVA-FITC uptake by cells from LPS-treated mice; solid histogram represents background FITC level for each subset from animals receiving OVA alone (no FITC). 1911

Discussion We show here that LPS has the ability to down-regulate surface expression of CD11c and CD11b on cdc subsets in the spleen. CD11c/CD18 and CD11b/CD18 are integrins expressed on the cell surface involved in cell adhesion and migration. They also bind components of the complement system and are therefore involved in pathogen sensing [7]. Furthermore, there are reports to suggest that CD11b/CD18 forms part of a CD14-mediated cell surface cluster in response to LPS [8, 9], and that CD11b/CD18 and CD11c/CD18 can induce signalling following LPS treatment independently of CD14 [10, 11]. Thus, down-regulation of CD11c and CD11b is probably a direct effect of LPS acting through the CD11b/CD11c binding, as well as a reflection of an LPS-induced shift in cellular function. Our observation of down-regulation of CD11c under inflammatory conditions is consistent with a recent report that CD11c on BMderived DCs cultured in vitro is down-regulated following TLR activation, as occurs with LPS treatment [12]. This could reflect preparation of the cells for migration from the spleen to the periphery following recognition of a danger signal. Investigation of cellular migration was not in the scope of this paper; however, LPS-induced migration of cdc subsets from the spleen may be accompanied by replacement with mature BM precursors, consistent with no observed perturbation of cell numbers in spleen after LPS treatment. The same paper reported an increase in MHC-II expression following Toll-like receptor activation. Our results confirm this observation following an in vivo challenge with LPS, an outcome that would improve the antigen-presenting capability of cells following activation. Relative proportions of mature dendritic and myeloid cells in spleen were not affected by LPS treatment; however, the proportional representation of p-predcs was lower in LPS-treated mice. As p- predcs represent an immature, inactivated form of pdcs, a drop in their numbers could reflect migration from the spleen to other organs. To test whether pdcs had migrated to other sites, we investigated mesenteric lymph nodes and saw no increase in numbers of pdcs. However, we did not progress to determine their presence in other lymph nodes or blood. The assumption is that changes in their migratory capacity may have led to dissemination of cells as a result of LPS treatment. Results presented here have important implications for the in vivo study of the innate immune response to infection, particularly if individual subsets responsible for a certain response are of interest. As we have shown, in the steady-state, splenic APC subsets are readily identifiable based on CD11b and CD11c expression, and can further be defined by CD8 and MHC-II expression. Following exposure to LPS, however, expression of these key markers changes, probably as a representation of the altered function of the cells under inflammatory conditions. These changes result in difficulties in identifying individual APC subsets, a factor that should be considered during analysis of an in vivo response to infection. Acknowledgements Funding for this project was provided by National Health and Medical Research Foundation of Australia to HO (Project grant #585443). Conflicts of interest The authors declare no conflict of interest. References 1. Merad M, Sathe P, Helft J, et al. The dendritic cell lineage: ontogeny and function of dendritic cells and their subsets in the steady state and the inflamed setting. Annu Rev Immunol. 2013; 31: 563 604. 2. Gahmberg CG, Tolvanen M, Kotovuori P. Leukocyte adhesion structure and function of human leukocyte beta2-integrins and their cellular ligands. Eur J Biochem. 1997; 245: 215 32. 3. Solovjov DA, Pluskota E, Plow EF. Distinct roles for the alpha and beta subunits in the functions of integrin alphambeta2. J Biol Chem. 2005; 280: 1336 45. 4. Tan JK, Quah BJ, Griffiths KL, et al. Identification of a novel antigen cross-presenting cell type in spleen. J Cell Mol Med. 2011; 15: 1189 99. 5. Sandor N, Kristof K, Parej K, et al. CR3 is the dominant phagocytotic complement receptor on human dendritic cells. Immunobiol. 2013; 218: 652 63. 6. Pfeiffer A, Bottcher A, Orso E, et al. Lipopolysaccharide and ceramide docking to CD14 provokes ligand-specific receptor clustering in rafts. Eur J Immunol. 2001; 31: 3153 64. 7. Scott MJ, Billiar TR. Beta2-integrin-induced p38 MAPK activation is a key mediator in the CD14/TLR4/MD2-dependent uptake of lipopolysaccharide by hepatocytes. J Biol Chem. 2008; 283: 29433 46. 8. Flaherty SF, Golenbock DT, Milham FH, et al. CD11/CD18 leukocyte integrins: new signaling receptors for bacterial endotoxin. J Surg Res. 1997; 73: 85 9. 9. Ingalls RR, Golenbock DT. CD11c/CD18, a transmembrane signaling receptor for lipopolysaccharide. J Exp Med. 1995; 181: 1473 9. 10. Singh-Jasuja H, Thiolat A, Ribon M, et al. The mouse dendritic cell marker CD11c is down-regulated upon cell activation through Toll-like receptor triggering. Immunobiol. 2013; 218: 28 39. 11. Schmidt A, Caron E, Hall A. Lipopolysaccharide-induced activation of beta2-integrin function in macrophages requires Irak kinase activity, p38 mitogen- activated protein kinase, and the Rap1 GTPase. Mol Cell Biol. 2001; 21: 438 48. 12. Morland B, Kaplan G. Macrophage activation in vivo and in vitro. Exp Cell Res. 1977; 108: 279 88. 1912