Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Tissue-specific expression of IgG receptors by human macrophages ex vivo

  • Christine W. Bruggeman ,

    Roles Formal analysis, Investigation, Methodology, Writing – original draft

    c.bruggeman@sanquin.nl

    Affiliation Department of Blood Cell Research, Sanquin Research and Landsteiner Laboratory, Academic Medical Center (AMC), University of Amsterdam, Amsterdam, The Netherlands

  • Julia Houtzager,

    Roles Resources

    Affiliation Department of Experimental Surgery, Academic Medical Center (AMC), University of Amsterdam, Amsterdam, The Netherlands

  • Barbara Dierdorp,

    Roles Resources

    Affiliation Department of Experimental Immunology, Academic Medical Center (AMC), University of Amsterdam, Amsterdam, The Netherlands

  • Jesper Kers,

    Roles Resources

    Affiliations Department of Pathology, Academic Medical Center (AMC), University of Amsterdam, Amsterdam, The Netherlands, Van 't Hoff Institute for Molecular Sciences, University of Amsterdam, Amsterdam, The Netherlands

  • Steven T. Pals,

    Roles Resources

    Affiliation Department of Pathology, Academic Medical Center (AMC), University of Amsterdam, Amsterdam, The Netherlands

  • René Lutter,

    Roles Resources

    Affiliation Department of Experimental Immunology, Academic Medical Center (AMC), University of Amsterdam, Amsterdam, The Netherlands

  • Thomas van Gulik,

    Roles Resources

    Affiliation Department of Experimental Surgery, Academic Medical Center (AMC), University of Amsterdam, Amsterdam, The Netherlands

  • Joke M. M. den Haan,

    Roles Supervision, Writing – review & editing

    Affiliation Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, The Netherlands

  • Timo K. van den Berg,

    Roles Supervision, Writing – review & editing

    Affiliation Department of Blood Cell Research, Sanquin Research and Landsteiner Laboratory, Academic Medical Center (AMC), University of Amsterdam, Amsterdam, The Netherlands

  • Robin van Bruggen,

    Roles Supervision, Writing – review & editing

    Affiliation Department of Blood Cell Research, Sanquin Research and Landsteiner Laboratory, Academic Medical Center (AMC), University of Amsterdam, Amsterdam, The Netherlands

  • Taco W. Kuijpers

    Roles Methodology, Supervision, Writing – review & editing

    Affiliations Department of Blood Cell Research, Sanquin Research and Landsteiner Laboratory, Academic Medical Center (AMC), University of Amsterdam, Amsterdam, The Netherlands, Emma Children's Hospital, Academic Medical Center (AMC), University of Amsterdam, Amsterdam, The Netherlands

Abstract

Recently it was discovered that tissue-resident macrophages derive from embryonic precursors, not only from peripheral blood monocytes, and maintain themselves by self-renewal. Most in-vitro studies on macrophage biology make use of in-vitro cultured human monocyte-derived macrophages. Phagocytosis of IgG-opsonized particles by tissue-resident macrophages takes place via interaction with IgG receptors, the Fc-gamma receptors (FcγRs). We investigated the FcγR expression on macrophages both in-vivo and ex-vivo from different human tissues. Upon isolation of primary human macrophages from bone marrow, spleen, liver and lung, we observed that macrophages from all studied tissues expressed high levels of FcγRIII, which was in direct contrast with the low expression on blood monocyte-derived macrophages. Expression levels of FcγRI were highly variable, with bone marrow macrophages showing the lowest and alveolar macrophages the highest expression. Kupffer cells in the liver were the only tissue-resident macrophages that expressed the inhibitory IgG receptor, FcγRIIB. This inhibitory receptor was also found to be expressed by sinusoidal endothelial cells in the liver. In sum, our immunofluorescence data combined with ex-vivo stainings of isolated macrophages indicated that tissue-resident macrophages are remarkably unique and different from monocyte-derived macrophages in their phenotypic expression of IgG receptors. Tissue macrophages show distinct tissue-specific FcγR expression patterns.

Introduction

Since the 1960s the dogma has been that the homeostasis of tissue-resident macrophages relies on the constant recruitment of blood monocytes [1]. Because of this assumption, most functional studies on macrophages have been performed on macrophages that have been in vitro cultured from peripheral blood monocytes. These monocyte-derived macrophages (MDMs) can be cultured with granulocyte macrophage-colony stimulating factor (GM-CSF) or macrophage-colony stimulating factor (M-CSF), generating the so-called M1 and M2 or pro-inflammatory and anti-inflammatory macrophages [2,3].

Recently, studies in rodents have shown that adult tissue macrophages are primarily derived from embryonic precursors that seed the tissues prior to birth, not from blood monocytes. These tissue-resident macrophages play a central role in homeostasis and maintain themselves by self-renewal [49]. Lung macrophages in the alveoli develop shortly after birth from hematopoietic stem cell-derived fetal monocytes and are not replaced by cells that derive from peripheral blood monocytes [10]. After birth, blood monocytes may indeed migrate into the tissues and differentiate into macrophages in case of inflammation [6]. Tissue-resident macrophages in the intestine and the skin are considered to be largely derived from blood monocytes, which can be explained by the fact that these sites are constantly exposed to the environment being subject to a low grade of mild inflammation. These intestinal MDMs contribute to homeostasis by having a high phagocytic and bactericidal activity, but they do not, in contrast to blood monocytes and other tissue macrophages, release proinflammatory cytokines upon phagocytosis [1113].

Macrophages play an important role in the phagocytosis of antibody-opsonized particles, a process that takes place via interaction with Fc-gamma receptors (FcγRs). These FcγRs, expressed by macrophages and other immune cells, recognize the constant region (Fc domain) of immunoglobulin G (IgG). The family of FcγRs consists of the high-affinity receptor FcγRI and the low-affinity receptors FcγRII and FcγRIII. All FcγRs, except for FcγRIIB and FcγRIIIB, are activating receptors because they contain an immunotyrosine-based activating motif (ITAM) or associate with the common ITAM-containing γ-chain [14]. MDMs that are cultured in the presence of GM-CSF or M-CSF have a distinctive FcγR expression pattern. GM-CSF macrophages, the so-called pro-inflammatory macrophages, mainly express FcγRI and FcγRIIIA, whereas M-CSF macrophages, the anti-inflammatory macrophages, predominantly express FcγRIIA [2,3]. Because macrophages are highly heterogeneous due to their origin and more in particular due to their tissue environment [10,15,16], we studied primary human macrophages in different tissues.

A detailed comparison of the FcγR expression patterns of macrophages from human tissues has not been available to date. For this reason, MDMs and different tissue-resident macrophages were compared by immunofluorescence stainings on tissue sections, and flow cytometry following isolation of macrophages from human bone marrow, spleen, liver and lung.

Methods

Human subjects

Peripheral blood from healthy volunteers was obtained in heparinized tubes. Informed consent was obtained from all volunteers. Spleen tissue was from organ transplant donors, obtained as part of the surgical procedure for HLA typing, as previously described [17,18]. Bone marrow was aspirated from the sternum of patients that underwent surgery for unrelated reasons and stored in heparinized tubes. Their hematopoietic compartment was considered to be healthy. Informed consent was obtained from all individuals. Alveolar macrophages were obtained by bronchoalveolar lavage (BAL) from patients suspected of COPD or sarcoidosis for diagnostic purposes. In case there was an excess of BAL fluid, cells were used for research in the present study. The alveolar macrophages may be in a more inflamed state than cells from completely healthy individuals. Pieces of lung tissue were obtained as part of surgical procedures and thereafter anonymized. Liver samples were from patients that underwent partial hepatectomy. The representative tissue samples were obtained from organ sections that were not involved in the primary disease for which surgery had been originally planned. Materials were collected as part of the diagnostic work-up and anonymously provided by the responsible pathologist (resident), in accordance with the Dutch law regarding the use of rest material for research purposes. The study was approved by the Medical Ethics Committee of the Academic Medical Center and Sanquin in Amsterdam and was performed in accordance with the Declaration of Helsinki.

Immunofluorescence

Small pieces of human spleen, liver or lung were embedded in Tissue Tek, frozen in liquid nitrogen vapor and stored at -80°C. Cryostat sections of 7 μm thickness were cut, fixed in acetone for 10 minutes and rehydrated in PBS. The sections were blocked with 5% human serum (Brocacef, Amsterdam, the Netherlands) in PBS for 15 minutes prior to 30 minutes staining with the following antibodies: anti-CD68 (clone Y1/82A, BD BioSciences), anti-VE-Cadherin (clone 55-7H1, BD BioSciences), anti-CD163 (clone MAC2-158, Trillium Diagnostics), anti-CD19 (clone HIB19, BioLegend), anti-CD64 (clone 10.1, BioLegend), anti-CD32a,b,c (clone AT10, Serotec), anti-CD32a (clone IV.3, SanBio), anti-CD32b,c (clone 2B6, a generous gift from MacroGenics, Rockville, MD) and anti-CD16 (clone 3G8, BioLegend). After washing with PBS, the sections were embedded in mowiol (Calbiochem). Isotype-matched antibodies were used as negative controls, i.e. the detection limit was increased until the level was reached at which the isotype control did not show any fluorescent signal anymore [18]. Stainings were analyzed using a DM6000 Leica immunofluorescence microscope or Leica SP8 confocal microscope.

Cell isolation

Splenocytes were isolated as previously described [18]. In short, a single cell suspension of splenocytes was prepared by injecting a piece of spleen with a collagenase buffer, containing DNAse, aggrastat and glucose. After filtering the suspension, erythrocytes were removed by incubating with an ice-cold lysis buffer. Bone marrow was processed by lysing the erythrocytes twice with an ice-cold isotonic ammonium chloride lysis buffer, for five minutes. Cells from BAL fluid were spun down by centrifuging for 10 minutes at 267g, 4°C. Cells were subsequently washed in PBS containing 0.5% bovine serum albumin (BSA) and 0.2 ppm K-EDTA and used for further analysis. To obtain a single cell suspension of liver cells, cells were isolated as previously described in detail [19]. In short, a piece of liver tissue was rinsed with perfusion solution to flush out the blood, and subsequently incubated with a collagenase buffer. The non-parenchymal cells were separated from the hepatocytes by low-speed centrifugation at gradually increasing rates. The non-parenchymal cells, including Kupffer cells, were used for further analysis.

All samples were processed on the day of collection, except for spleen tissue which was usually one day old. Cytospins were made and stained with May-Grünwald Giemsa for morphological analysis. To analyze the expression pattern of FcγRs, cells were analyzed by flow cytometry (see below).

Monocyte isolation and differentiation into monocyte-derived macrophages

Monocytes were isolated from peripheral blood mononuclear cells (PBMCs) and cultured into MDMs, essentially as described previously [2,3]. In short, monocytes were isolated by CD14 magnetic-activated cell separation (MACS) isolation kit (Miltenyi Biotec), according to manufacturer’s description. The isolated monocytes were then cultured for nine days in IMDM, supplemented with 10% FCS, L-glutamine and antibiotics, containing 10 ng/mL granulocyte macrophage-colony stimulating factor (GM-CSF) or 50 ng/mL macrophage-colony stimulating factor (M-CSF).

CD16pos and CD16neg monocytes were isolated by a two-step MACS isolation. First, platelet rich plasma was removed, after which monocytes were isolated from PBMCs by a negative isolation using the Pan monocyte isolation kit (Miltenyi Biotec), according to manufacturer’s description. Second, a positive isolation using the CD16 MACS isolation kit (Miltenyi Biotec) was used to separate the CD16pos and CD16neg monocytes. The cells were cultured in supplemented medium as described above.

Phenotyping and genotyping

Cells isolated from the various tissues were first gated based on canonical forward-sideward scatter and subsequently on autofluorescence, in a channel for which no staining was added. Macrophages were subsequently gated based on CD163 expression, using PE-labeled anti-CD163 (clone MAC2-158; Trillium Diagnostics), as we have previously published in detail [18]. For determining FcγR surface expression the following antibodies were used: FITC-labeled anti-CD64 (clone 10.1; BD Pharmingen), anti-CD32a,b,c (clone AT10; AbD Serotec), anti-CD32a (clone IV.3; Stem Cell Technologies), anti-CD32b,c (clone 2B6) and anti-CD16 (clone 3G8; BD Pharmingen). Stainings were performed in PBS containing 1% human serum albumin and 0.01% sodium azide. Proper FITC-labeled isotype controls were taken along. Samples were measured on a FACS CANTO II (BD BioSciences). We adhered to the guidelines for the use of flow cytometry and cell sorting in immunological studies.

All individuals were genotyped for the FCGR2/3 locus. Genotyping was performed as previously described [2022].

Statistical analysis

Statistical analysis was performed using GraphPad Prism 7.04. For comparison of FcγR expression levels between macrophages from the various tissues, Kruskal-Wallis tests were used, correcting for multiple comparisons using Dunn’s.

Results

Immunofluorescence of tissue sections

To investigate the FcγR expression pattern of tissue-resident macrophages, we performed co-stainings of macrophage markers and MoAbs specifically recognizing the different FcγR isoforms on sections of different tissues. Our study should be viewed with some limitations, because the different tissue samples were obtained from patients with various pathologies which were assumed to have no impact on FcγR expression, as indicated in the Methods section. Moreover, the tested tissues were not obtained from the same individual. Differences among individuals can thus not be excluded. We characterized human spleen red pulp macrophages by CD163 expression (Fig 1A). These macrophages expressed FcγRIIA and FcγRIIIA, but hardly if any FcγRI and FcγRIIB (Fig 1B–1E), as we have previously shown [18]. Marginal zone macrophages, characterized by the expression of CD169, were found to only express FcγRIIA [18].

thumbnail
Fig 1. Red pulp macrophages express FcγRIIA, and FcγRIII.

Immunofluorescence stainings of spleen sections with CD163 and (A) CD68 and CD19; (B) FcγRI and FcγRIIB/C; (C) FcγRII; (D) FcγRIIA and FcγRIIB/C (E) FcγRIII and CD19. Magnification 10x. Scale bars equal 100 μm. Figures are representative of n = 3 spleens from different donors. We have previously shown immunofluorescent stainings of human spleen tissue.18 Images in this figure are from sections from a different human spleen.

https://doi.org/10.1371/journal.pone.0223264.g001

Immunostainings of liver sections showed that Kupffer cells, stained by the expression of the common macrophage marker CD68, were also positive for CD163 (Fig 2A). Subsequent stainings revealed that Kupffer cells express FcγRII (Fig 2D and 2E), FcγRIII (Fig 2F), and no if any FcγRI (Fig 2C). Apart from being positive on Kupffer cells, liver sections showed remarkably high expression of FcγRIIB on tissue cells that were clearly different from macrophages and enriched throughout the liver sections. These cells could be morphologically characterized as sinusoidal endothelial cells. Co-stainings of anti-VE-Cadherin, which marks endothelial cells, and MoAb 2B6, specifically recognizing FcγRIIB [22], confirmed the expression of FcγRIIB on sinusoidal endothelial cells (Fig 2B).

thumbnail
Fig 2. Kupffer cells express FcγRIIA, FcγRIIB and FcγRIII.

Immunofluorescence stainings of liver sections with CD163 and (A) CD68 and FcγRIIB/C; (B) VECadherin and FcγRIIB/C; (C) FcγRI; (D) FcγRII; (E) FcγRIIA and FcγRIIB/C; (F) FcγRIII. Magnification 20x. Scale bars equal 50 μm. Figures are representative of n = 2 livers from different donors.

https://doi.org/10.1371/journal.pone.0223264.g002

Lung sections were found to be highly autofluorescent. Immunostainings of lung sections revealed that alveolar macrophages were also characterized by the expression of CD163 and the common macrophage marker CD68 (Fig 3A). Stainings showed that alveolar macrophages expressed FcγRI (Fig 3B) and FcγRIIA (Fig 3C and 3D). Based on these tissue sections, the expression of FcγRIII seemed to be low on alveolar macrophages (Fig 3E).

thumbnail
Fig 3. Lung macrophages express FcγRI and FcγRII.

Immunofluorescence stainings of lung sections with CD163, Hoechst and (A) CD68; (B) VECadherin; (C) FcγRI; (D) FcγRII; (E) FcγRIIA; (F) FcγRIII. Magnification 20x. Scale bars equal 50 μm. Figures are representative of n = 3 lungs from different donors. In (A) alveolar spaces are indicated with grey dashed lines.

https://doi.org/10.1371/journal.pone.0223264.g003

Ex vivo stainings of isolated macrophages

Because of the lack of accurate quantitation of FcγR expression and unavoidable background staining and autofluorescence, it is difficult to characterize the different tissue-resident macrophages in full detail based on immunostainings of tissue sections alone. For this reason and in order to compare the FcγR expression levels of macrophages from the different tissues with each other, we freshly isolated macrophages from the various tissues. By isolating the various macrophage subsets, we were able to ex vivo characterize these cells by cytospin and flow cytometry. Alveolar macrophages were obtained via a broncho-alveolar lavage and in this way, we studied these cells in more detail ex vivo by direct staining.

By cytospin we were able to analyze the morphology of the cells. Macrophages were easily detected by their round nucleus and granular bodies (S1 Fig). Macrophages that were cultured from blood monocytes in the presence of either GM-CSF or M-CSF were observed to be slightly bigger than tissue-resident macrophages by flow cytometry.

The isolated tissue-resident macrophages were further characterized by flow cytometry to determine the FcγR expression pattern of MDMs and macrophages from the various tissues available, i.e. bone marrow, spleen, liver and lung (Fig 4; Tables 1 and 2; S2 Fig). We always found homogeneous expression of the different tested FcγRs for macrophages from a certain tissue, suggesting that they form a homogeneous cell population and do not represent macrophage subsets (S3 Fig). MDMs were cultured in the presence of either GM-CSF or M-CSF for nine days and expressed an FcγR pattern distinct from each other. GM-CSF macrophages mainly expressed FcγRI and FcγRIIIA, whereas M-CSF macrophages expressed more FcγRIIA, as we have previously published [2,3].

thumbnail
Fig 4. FcγR expression by macrophages from different tissues.

Expression of (A) FcγRI, (B) FcγRIIA (MoAb IV.3), (C) FcγRIIB/C (MoAb 2B6), individuals encoding FCGR2C-ORF are indicated in red, and (D) FcγRIII by monocyte-derived macrophages cultured in the presence of GM-CSF, n = 9–32, and M-CSF, n = 9–39, by bone marrow macrophages, n = 14, red pulp macrophages, n = 83, Kupffer cells, n = 5, and alveolar macrophages, n = 5, determined by flow cytometry. For the cultured macrophages, individuals encoding FCGR2C-ORF were excluded from analysis. Data shown are median fluorescence intensity, corrected for the proper isotype control. For FcγRIIB/C expression, the levels on BM macrophages versus Kupffer cells from liver were nearly significant, p = 0.058. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001, ns = not significant.

https://doi.org/10.1371/journal.pone.0223264.g004

thumbnail
Table 1. Means and interquartile range (IQR) of FcγR expression levels by MDMs and tissue-resident macrophages, as shown in Fig 4A–4D.

https://doi.org/10.1371/journal.pone.0223264.t001

thumbnail
Table 2. Overview of FcγR expression by macrophages from BM, spleen, liver and lung.

https://doi.org/10.1371/journal.pone.0223264.t002

Bone marrow macrophages expressed no if any FcγRI (Fig 4A). Spleen and liver macrophages expressed a little more FcγRI, whereas alveolar macrophages expressed remarkably high levels of this receptor. FcγRI expression by alveolar macrophages reached expression levels that exceeded those of cultured macrophages. These findings were in line with the tissue section stainings, where alveolar macrophages were clearly shown to express FcγRI.

The expression of FcγRII was determined by staining with MoAb AT10, an antibody that is known to bind to all three isoforms FcγRIIA,B,C [22] (S2 Fig), whereas MoAb IV.3 most specifically recognizes FcγRIIA (Fig 4B). These stainings showed that FcγRIIA is expressed by macrophages from all different tissues. The expression of FcγRIIB, the only inhibitory IgG receptor, was specifically assessed by MoAb 2B6, which recognizes FcγRIIB/C [22]. By genotyping we determined which individuals have an FCGR2C-Open Reading Frame (ORF) and could therefore express FcγRIIC [20,21]. The presence of FCGR2C-ORF is indicated in red (Fig 4C). In case of an FCGR2C-STOP (indicated in black), no functional FcγRIIC can be expressed, in which case MoAb 2B6 specifically stains FcγRIIB. The inhibitory IgG receptor FcγRIIB was found to be expressed at a very low level if at all by most tissue macrophages except the Kupffer cells in the liver which–instead–demonstrated a significantly higher expression of FcγRIIB (Fig 4C).

Most remarkably, all tissue-resident macrophages, in contrast to MDMs, expressed FcγRIII (Fig 4D), at significantly higher levels than MDMs. We previously found the FcγRIII expression by red pulp macrophages to be specifically FcγRIIIA [18]. The expression level of FcγRIIIA correlates with FCGR3A copy number. For FCGR1 and FCGR2B no copy number variation occurs. This can explain the bigger spread in the FcγRIIIA levels compared to FcγRI and FcγRIIB. A subpopulation of about 10% of the blood monocytes is known to express FcγRIIIA (CD16). These CD16pos cells are believed to represent a specific subset of monocytes with a more mature phenotype than the CD16neg monocytes [23,24]. Reasoning that these specific circulating monocytes could be a source of a fraction of the tissue-resident macrophages, we investigated whether these cells showed a different phenotype upon the standard nine days of culturing in either GM-CSF or M-CSF. Upon culturing into MDMs, the MACS sorted peripheral blood monocytes being either CD16pos or CD16neg, expressed equal levels of this IgG receptor FcγRIIIA and of any of the other FcγRs (S4 Fig), which was already the case from day 3 of culturing. To exclude any specific effect of cell-cell interaction, we performed co-cultures of CD16pos and CD16neg monocytes and found the subsets of monocytes to form a homogeneous population.

Discussion

Recently parabiotic and adoptive-transplant studies in rodents have shown that tissue-resident macrophages are derived from embryonic precursors and maintain themselves by self-renewal [6,16]. Most tissue-resident macrophages, except those in the intestine and skin, do not differentiate from peripheral blood monocytes, but are derived from non-hematopoietic embryonic precursors [48]. Whereas most functional studies about human macrophages are being performed with in vitro cultured MDMs, the M1 and M2 cultured macrophages represent extremes of a multidimensional spectrum [9,10,25]. To investigate in vivo human tissue-resident macrophages and compare these with MDMs, we isolated primary human macrophages from various tissues. Since macrophages play an important role in the uptake of immune complexes and show phagocytosis of IgG-opsonized particles via their FcγRs, we investigated the tissue-specific IgG receptor expression of human macrophages in detail.

Tissue-resident macrophages showed a different FcγR expression profile compared to MDMs. The biggest difference was found to consist of high expression of FcγRIII, CD16, which was much higher in case of tissue-resident macrophages of all tested tissues than for MDMs.

A distinct subset of blood monocytes, around 10%, expresses CD16 [23]. Previous studies have shown that these cells are phenotypically and functionally different from the CD16neg monocytes and represent a more mature phenotype. For instance, phagocytosis of opsonized erythrocytes by CD16pos monocytes has been reported to be less efficient than by CD16neg monocytes [23,24]. It was also suggested that CD16pos monocytes would resemble certain types of tissue-resident macrophages [24]. Here we report that upon culturing the two subpopulations of monocytes into MDMs, these cells form a homogenous cell population that after nine days expressed equal levels of FcγRIII, although still at a much lower level than was observed in tissue-resident macrophages. Thus, the original difference in FcγRIII expression on circulating monocytes entirely disappeared once the cells were differentiated into MDMs.

A noticeable difference among macrophages from different tissues is their expression of FcγRI. FcγRI expression was very low if present on bone marrow macrophages, while spleen and liver macrophages expressed intermediate levels of FcγRI and its expression was very high on alveolar macrophages. This can be explained because FcγRI can be rapidly upregulated upon cellular activation by interferon-γ (IFNγ) and granulocyte (monocyte) colony-stimulating factor (G(M)-CSF) [26,27]. Upregulation of FcγRI on neutrophils is used as marker for bacterial infections and systemic inflammation [26,28]. We previously reported the positive correlation between increased FcγRI expression on splenic neutrophils and red pulp macrophages [18]. We hypothesize that bone marrow macrophages do not express FcγRI, because they are quiescent and non-activated, while alveolar macrophages are constantly exposed to environmental triggers [10], which could easily explain their high FcγRI expression level.

The function of FcγRI on alveolar macrophages remains to be investigated. Because FcγRI is a high-affinity IgG receptor, it is thought to be saturated by IgG in plasma which prevents the binding of immune complexes [29]. There is IgG present in alveoli too, although about 1000 times less than in plasma, as measured in human BAL fluids of healthy individuals which had an IgG concentration of around 9 μg/mL [30,31]. Since the IgG concentration in alveoli is much lower than in plasma, we hypothesize that FcγRI is partially unoccupied, making it a true high-affinity receptor for IgG-opsonized microbial agents. This is in line with recent publications that show that FcγRI is not fully saturated by IgG, neither in mice [32] nor in humans [33], making it readily available for immune complexes.

Another possibility is that FcγRI is internalized by other tissue-resident macrophages, because it is bound by monomeric IgG, and can be rapidly upregulated on the surface after stimulation by immune complexes [34]. Because of the lower IgG concentration in alveoli, alveolar macrophages may not have their FcγRI internalized and for that reason have increased surface expression, but this explanation seems unlikely, since we could not detect FcγRI in red pulp macrophages or Kupffer cells which would be expected even when internalized. Moreover, RNA levels of FCGR1 transcripts in red pulp macrophages are low, as we previously published [18], indicating that it is truly low expression and not due to immunofluorescence staining issues.

Another remarkable finding was that FcγRIIB, the only inhibitory receptor of the FcγR family, was highly expressed on sinusoidal endothelial cells in the liver. Previously, expression of FcγRIIB on sinusoidal endothelial cells was shown in mice and rats [3539], but to the best of our knowledge this has never been clearly shown for humans. Whereas large immune complexes, consisting for example of antibody-opsonized bacteria or blood cells, were shown to be cleared by mononuclear phagocytes, small immune complexes were cleared by liver sinusoidal endothelial cells via the interaction with FcγRIIB [35]. Removal of immune complexes from the circulation by the inhibitory FcγRIIB may well be able to prevent the inflammatory consequences of their presence in the tissue upon spillage from the gut drained through the portal vein into the liver. Since the liver receives blood via the portal vein, which transports blood from the intestine, it is known to be continuously exposed to endotoxin [40]. The liver must dampen the inflammatory reaction in order to prevent systemic immune activation [16,41]. In this way the coexistence of Kupffer cells and sinusoidal endothelial cells in the portal sinusoids, their unique expression profile of FcγRs and their contribution to the clearance of immune complexes all contribute to homeostasis.

As mentioned in the Methods section, spleen and liver tissues were obtained from individuals undergoing surgery for unrelated reasons. Their hematopoietic compartment was therefore considered to be healthy. The alveolar macrophages were obtained by BAL for diagnostic purposes from patients suspected of COPD or sarcoidosis. These cells may be in a more inflamed state than cells from healthy individuals. We cannot exclude this may influence the FcγR expression levels. Moreover, it cannot be excluded that fixation of the tissue could influence FcγR staining efficacy to different degrees in different tissues.

Our study is a unique description of the FcγR expression pattern of primary macrophages in-situ and following their purification from different human tissues and–as far as we know–the first of its kind. Future functional experiments will demand more material to isolate enough cells to show the contribution of the different FcγRs in binding and uptake of IgG-opsonized particles.

Recent studies made use of transgenic or knock-in mice that express human FcγRs and studied the FcγR expression pattern of different cell types in blood and various tissues [42,43], including tumor-associated macrophages [44]. The FcγR expression pattern of blood leukocytes from these mice were compared to that of leukocytes from human blood. Although the FcγR expression patterns look similar between species, they are not identical. For instance, a lower FcγRIIA expression on mouse neutrophils and the presence of FcγRIIB on mouse monocytes, while this receptor is not expressed by human monocytes. This indicates that, although informative, FcγR expression by tissue macrophages obtained with transgenic or knock-in mice expressing human FcγRs, cannot be directly translated to humans.

The differences in FcγR expression profile between MDMs and tissue-resident macrophages, but also the differences between macrophages from the various tissues, raise the question whether these differences are due to the different origin of these cells or due to their milieu [16,45]. A recent study by Lavin et al. showed that the chromatin state of tissue-resident macrophages can partially be explained by their origin and partially by their environment. It is a combination of lineage-specific and tissue-specific factors that determines the identity and plasticity of tissue-resident macrophages [46]. Our study shows the remarkable differences between macrophages from different tissues and thereby highlights the diversity and complexity of this cell type.

In sum, we here show for the first time that human macrophages from bone marrow, spleen, liver and lung have unique FcγR expression patterns. We determined the expression patterns by in vivo characterization of stainings of tissue sections. Upon isolating macrophages for ex vivo characterization we investigated these cells in full detail. We showed that splenic red pulp macrophages express FcγRIIA and FcγRIII, Kupffer cells from the liver express FcγRIIA, FcγRIIB and FcγRIII. FcγRI was sometimes expressed to low extent. Alveolar macrophages express FcγRI, FcγRIIA and FcγRIII. We found the FcγR expression patterns of tissue-resident macrophages to be totally different from the expression patterns of in vitro cultured MDMs.

Supporting information

S1 Fig. Cytospins of macrophages.

Cytospins of monocyte-derived macrophages cultured in the presence of (A) GM-CSF and (B) M-CSF, cytospins of tissue macrophages: (C) bone marrow, (D) sorted red pulp macrophages, (E) cells from liver and (F) cells from bronchoalveolar lavage. Magnification 50x. Figures are representative of at least n = 3 different donors.

https://doi.org/10.1371/journal.pone.0223264.s001

(TIF)

S2 Fig. FcγRII expression by macrophages from different tissues.

Expression of FcγRIIA,B,C (MoAb AT10) by monocyte-derived macrophages cultured in the presence of GM-CSF, n = 32, and M-CSF, n = 39, by bone marrow macrophages, n = 14, red pulp macrophages, n = 83, Kupffer cells, n = 5, and alveolar macrophages, n = 5, determined by flow cytometry. Individuals encoding FCGR2C-ORF are indicated in red. Data shown are median fluorescence intensity (MFI), corrected for the proper isotype control (ΔMFI), the error bars represent SEM. *p < 0.05, **p < 0.01, ns = not significant.

https://doi.org/10.1371/journal.pone.0223264.s002

(TIF)

S3 Fig. FcγR expression of macrophages from human spleen show homogeneous cell populations.

Flow cytometry stainings of anti-FcγRI (clone 10.1), anti-FcγRIIA (clone IV.3), anti-FcγRIIB (clone 2B6) and anti-FcγRIII (clone 3G8) of macrophages (upper panel) and monocytes (lower panel) isolated from human spleen and their appropriate isotype controls (upper rows). Median fluorescence intensity is shown on the x-axis. Data are from representative spleen sample.

https://doi.org/10.1371/journal.pone.0223264.s003

(TIF)

S4 Fig. Upon culturing into MDMs, the originally CD16pos and CD16neg monocytes show identical FcγR expression levels.

FcγR expression by MDMs cultured in the presence of GM-CSF (left) and M-CSF (right) of originally CD16pos monocytes (black) and CD16neg monocytes (grey), n = 4. Data shown are median fluorescence intensity (MFI), corrected for the proper isotype control (ΔMFI), error bars represent SEM.

https://doi.org/10.1371/journal.pone.0223264.s004

(TIF)

Acknowledgments

The authors would like to thank Mrs. Naneth Goes (Department of Blood Cell Research, Sanquin, Amsterdam, The Netherlands) and Mrs. Katarina Olesek (Department of Molecular Cell Biology and Immunology, VUmc, Amsterdam, The Netherlands) for assistance with the experiments.

References

  1. 1. van Furth R., and Cohn Z. A. 1968. The origin and kinetics of mononuclear phagocytes. J Exp Med 128: 415–435. pmid:5666958
  2. 2. Bruggeman C. W., Dekkers G., Bentlage A. E. H., Treffers L. W., Nagelkerke S. Q., Lissenberg-Thunnissen S., Koeleman C. A. M., Wuhrer M., van den Berg T. K., Rispens T., Vidarsson G., and Kuijpers T. W. 2017. Enhanced Effector Functions Due to Antibody Defucosylation Depend on the Effector Cell Fcgamma Receptor Profile. J Immunol 199: 204–211. pmid:28566370
  3. 3. Nagelkerke S. Q., Dekkers G., Kustiawan I., van de Bovenkamp F. S., Geissler J., Plomp R., Wuhrer M., Vidarsson G., Rispens T., van den Berg T. K., and Kuijpers T. W. 2014. Inhibition of FcgammaR-mediated phagocytosis by IVIg is independent of IgG-Fc sialylation and FcgammaRIIb in human macrophages. Blood 124: 3709–3718. pmid:25352126
  4. 4. Ginhoux F., and Jung S. 2014. Monocytes and macrophages: developmental pathways and tissue homeostasis. Nat Rev Immunol 14: 392–404. pmid:24854589
  5. 5. Gentek R., Molawi K., and Sieweke M. H. 2014. Tissue macrophage identity and self-renewal. Immunol Rev 262: 56–73. pmid:25319327
  6. 6. Hashimoto D., Chow A., Noizat C., Teo P., Beasley M. B., Leboeuf M., Becker C. D., See P., Price J., Lucas D., Greter M., Mortha A., Boyer S. W., Forsberg E. C., Tanaka M., van Rooijen N., Garcia-Sastre A., Stanley E. R., Ginhoux F., Frenette P. S., and Merad M. 2013. Tissue-resident macrophages self-maintain locally throughout adult life with minimal contribution from circulating monocytes. Immunity 38: 792–804. pmid:23601688
  7. 7. Ginhoux F., Greter M., Leboeuf M., Nandi S., See P., Gokhan S., Mehler M. F., Conway S. J., Ng L. G., Stanley E. R., Samokhvalov I. M., and Merad M. 2010. Fate mapping analysis reveals that adult microglia derive from primitive macrophages. Science 330: 841–845. pmid:20966214
  8. 8. Schulz C., Gomez Perdiguero E., Chorro L., Szabo-Rogers H., Cagnard N., Kierdorf K., Prinz M., Wu B., Jacobsen S. E., Pollard J. W., Frampton J., Liu K. J., and Geissmann F. 2012. A lineage of myeloid cells independent of Myb and hematopoietic stem cells. Science 336: 86–90. pmid:22442384
  9. 9. Gordon S., and Martinez-Pomares L. 2017. Physiological roles of macrophages. Pflugers Archiv: European journal of physiology 469: 365–374. pmid:28185068
  10. 10. Epelman S., Lavine K. J., and Randolph G. J. 2014. Origin and functions of tissue macrophages. Immunity 41: 21–35. pmid:25035951
  11. 11. Gordon S., and Taylor P. R. 2005. Monocyte and macrophage heterogeneity. Nat Rev Immunol 5: 953–964. pmid:16322748
  12. 12. Smythies L. E., Sellers M., Clements R. H., Mosteller-Barnum M., Meng G., Benjamin W. H., Orenstein J. M., and Smith P. D. 2005. Human intestinal macrophages display profound inflammatory anergy despite avid phagocytic and bacteriocidal activity. J Clin Invest 115: 66–75. pmid:15630445
  13. 13. Gonzalez-Dominguez E., Samaniego R., Flores-Sevilla J. L., Campos-Campos S. F., Gomez-Campos G., Salas A., Campos-Pena V., Corbi A. L., Sanchez-Mateos P., and Sanchez-Torres C. 2015. CD163L1 and CLEC5A discriminate subsets of human resident and inflammatory macrophages in vivo. J Leukoc Biol 98: 453–466. pmid:25877931
  14. 14. Bruhns P., Iannascoli B., England P., Mancardi D. A., Fernandez N., Jorieux S., and Daeron M. 2009. Specificity and affinity of human Fcgamma receptors and their polymorphic variants for human IgG subclasses. Blood 113: 3716–3725. pmid:19018092
  15. 15. Davies L. C., Jenkins S. J., Allen J. E., and Taylor P. R. 2013. Tissue-resident macrophages. Nat Immunol 14: 986–995. pmid:24048120
  16. 16. Varol C., Mildner A., and Jung S. 2015. Macrophages: development and tissue specialization. Annu Rev Immunol 33: 643–675. pmid:25861979
  17. 17. Nagelkerke S. Q., aan de Kerk D. J., Jansen M. H., van den Berg T. K., and Kuijpers T. W. 2014. Failure to detect functional neutrophil B helper cells in the human spleen. PLoS One 9: e88377. pmid:24523887
  18. 18. Nagelkerke S. Q., Bruggeman C. W., den Haan J. M. M., Mul E. P. J., van den Berg T. K., van Bruggen R., and Kuijpers T. W. 2018. Red pulp macrophages in the human spleen are a distinct cell population with a unique expression of Fc-gamma receptors. Blood advances 2: 941–953. pmid:29692344
  19. 19. Werner M., Driftmann S., Kleinehr K., Kaiser G. M., Mathe Z., Treckmann J. W., Paul A., Skibbe K., Timm J., Canbay A., Gerken G., Schlaak J. F., and Broering R. 2015. All-In-One: Advanced preparation of Human Parenchymal and Non-Parenchymal Liver Cells. PLoS One 10: e0138655. pmid:26407160
  20. 20. Breunis W. B., van Mirre E., Geissler J., Laddach N., Wolbink G., van der Schoot E., de Haas M., de Boer M., Roos D., and Kuijpers T. W. 2009. Copy number variation at the FCGR locus includes FCGR3A, FCGR2C and FCGR3B but not FCGR2A and FCGR2B. Hum Mutat 30: E640–650. pmid:19309690
  21. 21. Tsang A. S. M. W., Nagelkerke S. Q., Bultink I. E., Geissler J., Tanck M. W., Tacke C. E., Ellis J. A., Zenz W., Bijl M., Berden J. H., de Leeuw K., Derksen R. H., Kuijpers T. W., and Voskuyl A. E. 2016. Fc-gamma receptor polymorphisms differentially influence susceptibility to systemic lupus erythematosus and lupus nephritis. Rheumatology (Oxford) 55: 939–948.
  22. 22. van der Heijden J., Breunis W. B., Geissler J., de Boer M., van den Berg T. K., and Kuijpers T. W. 2012. Phenotypic variation in IgG receptors by nonclassical FCGR2C alleles. J Immunol 188: 1318–1324. pmid:22198951
  23. 23. Passlick B., Flieger D., and Ziegler-Heitbrock H. W. 1989. Identification and characterization of a novel monocyte subpopulation in human peripheral blood. Blood 74: 2527–2534. pmid:2478233
  24. 24. Ziegler-Heitbrock H. W., Fingerle G., Strobel M., Schraut W., Stelter F., Schutt C., Passlick B., and Pforte A. 1993. The novel subset of CD14+/CD16+ blood monocytes exhibits features of tissue macrophages. Eur J Immunol 23: 2053–2058. pmid:7690321
  25. 25. Ginhoux F., Schultze J. L., Murray P. J., Ochando J., and Biswas S. K. 2016. New insights into the multidimensional concept of macrophage ontogeny, activation and function. Nat Immunol 17: 34–40. pmid:26681460
  26. 26. Zhang H., Ling X. L., Wu Y. Y., Lu M. H., Guo H., Zhang P. B., Zhao X. Y., and Yang S. M. 2014. CD64 expression is increased in patients with severe acute pancreatitis: clinical significance. Gut Liver 8: 445–451. pmid:25071912
  27. 27. Gericke G. H., Ericson S. G., Pan L., Mills L. E., Guyre P. M., and Ely P. 1995. Mature polymorphonuclear leukocytes express high-affinity receptors for IgG (Fc gamma RI) after stimulation with granulocyte colony-stimulating factor (G-CSF). J Leukoc Biol 57: 455–461. pmid:7533820
  28. 28. Matt P., Lindqvist U., and Kleinau S. 2015. Up-regulation of CD64-expressing monocytes with impaired FcgammaR function reflects disease activity in polyarticular psoriatic arthritis. Scand J Rheumatol 44: 464–473. pmid:26084203
  29. 29. van der Poel C. E., Spaapen R. M., van de Winkel J. G., and Leusen J. H. 2011. Functional characteristics of the high affinity IgG receptor, FcgammaRI. J Immunol 186: 2699–2704. pmid:21325219
  30. 30. Peebles R. S. Jr., Liu M. C., Lichtenstein L. M., and Hamilton R. G. 1995. IgA, IgG and IgM quantification in bronchoalveolar lavage fluids from allergic rhinitics, allergic asthmatics, and normal subjects by monoclonal antibody-based immunoenzymetric assays. Journal of immunological methods 179: 77–86. pmid:7868927
  31. 31. Van Vyve T., Chanez P., Bernard A., Bousquet J., Godard P., Lauwerijs R., and Sibille Y. 1995. Protein content in bronchoalveolar lavage fluid of patients with asthma and control subjects. The Journal of allergy and clinical immunology 95: 60–68. pmid:7822665
  32. 32. Gillis C. M., Zenatti P. P., Mancardi D. A., Beutier H., Fiette L., Macdonald L. E., Murphy A. J., Celli S., Bousso P., Jonsson F., and Bruhns P. 2017. In vivo effector functions of high-affinity mouse IgG receptor FcgammaRI in disease and therapy models. Journal of autoimmunity 80: 95–102. pmid:27745779
  33. 33. Mancardi D. A., Albanesi M., Jonsson F., Iannascoli B., Van Rooijen N., Kang X., England P., Daeron M., and Bruhns P. 2013. The high-affinity human IgG receptor FcgammaRI (CD64) promotes IgG-mediated inflammation, anaphylaxis, and antitumor immunotherapy. Blood 121: 1563–1573. pmid:23293080
  34. 34. van der Poel C. E., Karssemeijer R. A., Boross P., van der Linden J. A., Blokland M., van de Winkel J. G., and Leusen J. H. 2010. Cytokine-induced immune complex binding to the high-affinity IgG receptor, FcgammaRI, in the presence of monomeric IgG. Blood 116: 5327–5333. pmid:20805361
  35. 35. Ganesan L. P., Kim J., Wu Y., Mohanty S., Phillips G. S., Birmingham D. J., Robinson J. M., and Anderson C. L. 2012. FcgammaRIIb on liver sinusoidal endothelium clears small immune complexes. J Immunol 189: 4981–4988. pmid:23053513
  36. 36. March S., Hui E. E., Underhill G. H., Khetani S., and Bhatia S. N. 2009. Microenvironmental regulation of the sinusoidal endothelial cell phenotype in vitro. Hepatology 50: 920–928. pmid:19585615
  37. 37. Mousavi S. A., Sporstol M., Fladeby C., Kjeken R., Barois N., and Berg T. 2007. Receptor-mediated endocytosis of immune complexes in rat liver sinusoidal endothelial cells is mediated by FcgammaRIIb2. Hepatology 46: 871–884. pmid:17680646
  38. 38. Ganesan L. P., Mohanty S., Kim J., Clark K. R., Robinson J. M., and Anderson C. L. 2011. Rapid and efficient clearance of blood-borne virus by liver sinusoidal endothelium. PLoS pathogens 7: e1002281. pmid:21980295
  39. 39. Mates J. M., Yao Z., Cheplowitz A. M., Suer O., Phillips G. S., Kwiek J. J., Rajaram M. V., Kim J., Robinson J. M., Ganesan L. P., and Anderson C. L. 2017. Mouse Liver Sinusoidal Endothelium Eliminates HIV-Like Particles from Blood at a Rate of 100 Million per Minute by a Second-Order Kinetic Process. Frontiers in immunology 8: 35. pmid:28167948
  40. 40. Laskin D. L., Weinberger B., and Laskin J. D. 2001. Functional heterogeneity in liver and lung macrophages. J Leukoc Biol 70: 163–170. pmid:11493607
  41. 41. Krenkel O., and Tacke F. 2017. Liver macrophages in tissue homeostasis and disease. Nat Rev Immunol 17: 306–321. pmid:28317925
  42. 42. Beutier H., Hechler B., Godon O., Wang Y., Gillis C. M., de Chaisemartin L., Gouel-Cheron A., Magnenat S., Macdonald L. E., Murphy A. J., Chollet-Martin S., Longrois D., Gachet C., Bruhns P., and Jonsson F. 2018. Platelets expressing IgG receptor FcgammaRIIA/CD32A determine the severity of experimental anaphylaxis. Science immunology 3.
  43. 43. Gillis C. M., Jonsson F., Mancardi D. A., Tu N., Beutier H., Van Rooijen N., Macdonald L. E., Murphy A. J., and Bruhns P. 2017. Mechanisms of anaphylaxis in human low-affinity IgG receptor locus knock-in mice. The Journal of allergy and clinical immunology 139: 1253–1265.e1214. pmid:27568081
  44. 44. Arce Vargas F., Furness A. J. S., Litchfield K., Joshi K., Rosenthal R., Ghorani E., Solomon I., Lesko M. H., Ruef N., Roddie C., Henry J. Y., Spain L., Ben Aissa A., Georgiou A., Wong Y. N. S., Smith M., Strauss D., Hayes A., Nicol D., O'Brien T., Martensson L., Ljungars A., Teige I., Frendeus B., Pule M., Marafioti T., Gore M., Larkin J., Turajlic S., Swanton C., Peggs K. S., and Quezada S. A. 2018. Fc Effector Function Contributes to the Activity of Human Anti-CTLA-4 Antibodies. Cancer cell 33: 649–663.e644. pmid:29576375
  45. 45. Gautier E. L., and Yvan-Charvet L. 2014. Understanding macrophage diversity at the ontogenic and transcriptomic levels. Immunol Rev 262: 85–95. pmid:25319329
  46. 46. Lavin Y., Winter D., Blecher-Gonen R., David E., Keren-Shaul H., Merad M., Jung S., and Amit I. 2014. Tissue-resident macrophage enhancer landscapes are shaped by the local microenvironment. Cell 159: 1312–1326. pmid:25480296