A dense network of macrophages and dendritic cells (DC) expressing the chemokine receptor CX3CR1 populates most tissues. We recently reported that CX3CR1 regulates the abundance of CD11c+ DC in the kidney and thereby promotes renal inflammation in glomerulonephritis. Given that chronic inflammation usually causes fibrosis, we hypothesized that CX3CR1 deficiency should attenuate renal fibrosis. However, when we tested this hypothesis using the DC-independent murine fibrosis model of unilateral ureteral obstruction, kidney fibrosis was unexpectedly more severe, despite less intrarenal inflammation. Two-photon imaging and flow cytometry revealed in kidneys of CX3CR1-deficient mice more motile Ly6C/Gr-1+ macrophages. Flow cytometry verified that renal macrophages were more abundant in the absence of CX3CR1 and produced more of the key profibrotic mediator, TGF-β. Macrophages accumulated because of higher intrarenal proliferation, despite reduced monocyte recruitment and higher signs of apoptosis within the kidney. These findings support the theory that tissue macrophage numbers are regulated through local proliferation and identify CX3CR1 as a regulator of such proliferation. Thus, CX3CR1 inhibition should be avoided in DC-independent inflammatory diseases because it may promote fibrosis.

The renal mononuclear phagocyte system consists of an extensive network of mononuclear phagocytes (i.e., dendritic cells [DCs] and macrophages) (1, 2). Although there is no clear demarcation between these cell types, DCs are typically specialized at regulating other immune effector cells, especially T cells, whereas macrophages preferentially act as immune effectors in innate immune responses or contribute to tissue repair (36). In the healthy kidney, most resident immune cells display phenotypic and functional characteristics of DCs and can be distinguished from macrophages by expression of the CD11c molecule (7). Furthermore, CD11c+ kidney cells possess a transcriptome that resembles that of DCs but not of macrophages (8, 9). Although kidney DCs have been shown to perform sentinel and regulatory functions in inflammation and infection (10), macrophages contributed to host protection from infection and maintained tissue homeostasis (1, 11, 12). In contrast, inappropriate macrophage functions promoted fibrosis in various organs (13, 14). In the kidney, macrophages but not DCs mediated fibrosis in unilateral ureter ligation (UUO) and after ischemia/reperfusion (1518). Several mechanisms contribute to renal fibrosis, including the production of the main profibrotic molecule TGF-β (14, 19). In the kidney, TGF-β promotes fibrosis by activating fibroblasts and myofibroblasts, modulating the expression of tissue inhibitors of matrix metalloproteases and by directly promoting synthesis of collagens (20). TGF-β secretion has been linked to monocytes and macrophages (14), suggesting that macrophages might mediate kidney fibrosis through this mediator.

Tissue macrophages partially originate from embryonic progenitors of the yolk sac or the fetal liver (21, 22), and from infiltrating bone marrow–dependent progenitors such as monocytes, whereas DCs can develop from monocytes or pre-DCs (2325). The exact contribution of the different precursors to the renal mononuclear cell system is unclear. The chemokine receptor CX3CR1, also known as fractalkine receptor, is ubiquitously expressed on most tissue macrophages and DCs but does not play a major role for their ontogeny, homeostatic migration, or colonization of tissues with resident phagocytes (26, 27), except kidney DCs (3) and intestinal macrophages (2830). There is evidence that the survival of monocytes and tissue macrophages depends on CX3CR1 (31-33), and this required interaction with cell surface–bound CX3CL1 (34). CX3CL1+ cells have been shown to be present within glomeruli and in the tubular compartment (35), but their exact identity is unclear.

Under inflammatory conditions, CCR2-dependent monocytes expressing the markers Ly6C and Gr1 enter inflamed tissues (3638) and rapidly give rise to DCs and macrophages (31, 3941). Inflammatory Gr1+ macrophages possess profibrotic properties (42), and their depletion or genetic CCR2 deficiency attenuated tissue damage and fibrosis in UUO (16), lupus nephritis (43), and ischemia/reperfusion (44). Also, CX3CR1 has been implicated in monocyte recruitment into inflamed tissues, like arteriosclerotic vessels (45), the listeria-infected spleen (46), the eye (47), in colitis models (28), or the skin (48). Furthermore, CX3CR1 may exert profibrotic functions (4955).

DCs and macrophages of the kidney abundantly express CX3CR1 (56). CX3CR1 deficiency reduced entry of their progenitors into the kidney and attenuated symptoms and fibrosis in ischemia/reperfusion injury (44, 49), hypertensive kidney damage (54), and crescentic glomerulonephritis (3, 57). In contrast, the loss of renal phagocytes aggravated renal candidiasis (58) but not pyelonephritis (3). Also, studies on human biopsy material from fibrotic kidneys described upregulation of CX3CR1, supporting a pathogenic role (59). However, in toxin- and in obstruction-induced liver fibrosis, CX3CR1 was protective (50). The role of CX3CR1 in the obstruction-induced standard kidney fibrosis model, UUO, has not been clarified yet. In this study, we have addressed this open question, assuming that the marked loss of renal DCs in CX3CR1 deficiency would attenuate inflammation and fibrosis. However, we found that CX3CR1 was protective in UUO because of a novel inhibitory effect on the other CX3CR1-expressing mononuclear phagocyte type of the kidney, the macrophages.

Eight- to 14-wk-old Cd45.1, Cd11cEYFP (60), Cx3cr1GFP/+, and Cx3cr1GFP/GFP mice (61), all on a C57BL/6 background, were bred and kept under specific-pathogen-free condition at the animal facilities of the University of Bonn and Monash Medical Centre. Institutional and Government Review Boards have approved all animal studies.

Adult male mice were anesthetized by inhalation of isofluorane. UUO was performed through a midline incision. The left ureter was tied with silk suture at two points and permanently ligated. Sham-operated mice underwent an identical midline incision but without ureteric ligation. At 3, 5, and 7 d postobstruction, mice were sacrificed, and kidneys were collected.

Intravital multiphoton microscopy of the intact kidney was performed on mice 3 d post-UUO, anesthetized by i.p. injection of 150 mg/kg ketamine hydrochloride and 10 mg/kg xylazine, as described previously (18, 62). The jugular vein was cannulated, and the left kidney was exteriorized through a lateral incision, immersed in bicarbonate buffered saline at 37°C, and covered with a coverslip. Mice were kept at 37°C. Time lapse recordings of GFP+ cells in the kidney of Cx3cr1GFP/+ and Cx3cr1GFP/GFP mice were captured using a Leica SP5 mulitphoton microscope system (Leica Microsystems, Mannheim, Germany) with a ×20 1.0 NA water immersion objective (18, 63). Fluorophores were excited by a SpectraPhysics MaiTai pulsed infrared laser tuned to 860 nm, with emissions captured by non-descanned detectors with 525/50 nm (GFP), 585/40 nm (tetramethylrhodamine isothiocyanate), and 650/50 nm (Alexa 633) filters. Celltracker Orange CMRA (C34551; Molecular Probes) was used as a counterstain to provide visual context. Images were collected with ∼120 μm Z-depth in 6-μm steps, with one Z-stack every 30s for 20–30 min and analyzed using Imaris (version 7.0; Bitplane AG). To determine GFP+ cell motility, individual cells were tracked in vivo and a motile cell defined as one that had moved at least the diameter of the cell body. To determine cell activity, a morphology index was used as previously described (18), relating to the sphericity of cells described by the rate of change in morphology index (ΔMI). Cells with relatively low ΔMI values change morphology less than cells with higher ΔMI values.

Methyl Carnoyl’s fixed paraffin-embedded murine kidneys were sectioned at 1 μm and stained with periodic acid–Schiff’s (PAS) for evaluation of hydronephrosis and overall injury. For the evaluation of fibrosis, Sirius Red stained sections as well as α-smooth muscle actin (αSMA) immunohistochemistry were evaluated using computer-based morphometric analyses by a pathologist in blinded fashion as described previously (64). In short, for both stainings, the percentage of positively stained area was calculated from 20 cortical fields per section, representing nearly the whole cortex. Indirect immunoperoxidase procedure was performed as described previously (64). HRP coupled to human αSMA (clone 1A4; DakoCytomation, Glostrup, Denmark), which cross-react with murine αSMA was used. Fluorescence microscopy was performed on 5-μm paraffin sections from 4% PFA-fixed kidneys. GFP-expressing cells in Cx3cr1GFP/+ mice were identified by nuclei staining and quantified using the IX71 microscope and the CellR software (Olympus).

Kidneys were digested with collagenase and DNAse-I (both from Sigma-Aldrich) as described previously (7). Briefly, kidneys were digested for 45 min, and tubular fragments from digested kidneys were removed by filtration through a 100-μm nylon mesh.

After treatment with Fc-blocking Abs (2.4G2) cells were stained with fluorochrome-labeled mAbs against F4/80 (CI:A3-1), CD45 (30-F11), CD11c (N418), KI67 (SolA15; isotype control: eBR2a), Gr1 (RB6-8C5), and Annexin V (number 550474; BD Biosciences). For intracellular cytokine staining, single-cell suspensions were incubated in RPMI 1640 medium (10% FCS, penicillin/streptomycin, and glutamine) with 1 μl/ml GolgiPlug and 1 μl/ml GolgiStop (BD Biosciences) for 4 h at 37°C. After cell surface staining, cells were fixed in Cytofix (BD Biosciences) and permeabilized with PermWash (BD Biosciences). Intracellular staining was performed for 30 min at room temperature using fluorochrome-labeled mAbs against TGFβ (TW7-16B4; isotype control: MOPC-21). For analysis of proliferation, BrdU was administered 3 and 4 d after ligation, and incorporation of BrdU was analyzed on day 5 after ligation by flow cytometry (number 557892; BD Biosciences). Cells were analyzed with a BD Canto II and a BD Fortessa (BD Biosciences), and data were analyzed using Flowjo (Tree Star). For concatenated data presentation, data from all mice within the same group was combined using Flowjo.

Single-cell suspensions from the bone marrows were prepared by flushing the bones with sterile and cold PBS. Bone marrow monocytes were enriched by excluding CD19+CD3+CD11c+ cells. Bone marrow cells (1–5 × 106) were transferred into mice.

The protein levels of cytokines in the supernatant of mechanically homogenized kidneys were determined by Luminex assay. The growth factors M-CSF and GM-CSF were determined by using ELISA (R&D Systems). All other levels were determined by a flow cytometry–based bead assay (eBioscience).

Whole-tissue RNA from kidneys was extracted using NucleoSpin RNA II kit (Macherey-Nagel), according to the manufacturer’s instructions. RNA was reverse transcribed into cDNA using the High-Capacity Reverse Transcription Kit (Applied Biosystems). Quantitative PCR was performed for 40 cycles using SYBR Green (Applied Biosystems). QuantiTect Primer assays (Qiagen) were used to detect mouse gene expression of Stat1 (QT00162183), Stat6 (QT01042503), Arg1 (QT00134288), Hmox (QT00159915), Irf5 (QT00252623), Retnla (QT00254359), and Mrc1 (QT00103012). All samples were run in duplicates and normalized to Hprt primers (Invitrogen), forward primer (5′-GTCCCAGCGTCGTGATTAGCGAT-3′) and reverse primer (5′-GGGCCACAATGTGATGGCCTCC-3′).

Results are expressed as mean ± SEM. Comparisons were drawn using parametric T-Test and nonparametric Mann–Whitney test (Prism 5; Graphpad Software, San Diego, CA). Significant p values were expressed as follows: *p < 0.05; **p < 0.01; ***p < 0.001.

The chemokine receptor CX3CR1 has been shown to promote renal inflammation in several models (3, 49, 54, 5759, 65). Given that chronic kidney inflammation often leads to fibrosis, we hypothesized that CX3CR1 deficiency should attenuate renal fibrosis. To test this hypothesis, we induced one of the standard kidney fibrosis models, UUO in mice expressing or lacking CX3CR1. We used Cx3cr1GFP/GFP mice that homozygously express a GFP reporter under the control of the CX3CR1 promoter and are consequently CX3CR1 deficient. As controls, we used Cx3cr1GFP/+ mice, which express the GFP reporter heterozygously and CX3CR1-competent wild-type mice (Cx3cr1+/+ mice). Surprisingly, kidney fibrosis in CX3CR1-deficient mice was notably aggravated compared with control mice 7 d after UUO, as evidenced by higher Sirius Red collagen staining in the ligated kidneys of Cx3cr1GFP/GFP mice (Fig. 1A, 1B). Also, αSMA staining for profibrotic myofibroblasts was stronger in ligated kidneys of CX3CR1-deficient mice (Fig. 1C, 1D). These findings revealed an unexpected antifibrotic role of CX3CR1 in renal fibrosis.

FIGURE 1.

Lack of CX3CR1 increases renal fibrosis in UUO. (AD) Fibrosis was quantified by Sirius red staining (A and B) and αSMA immunostaining (C and D) of sections of ligated or contralateral kidneys from CX3CR1-competent (Cx3cr1+/+,Cx3cr1GFP/+ [left images or white and light gray bars]) and CX3CR1-deficient (Cx3cr1GFP/GFP [right images or dark gray bars]) mice 7 d after ureter ligation. (A and C) Scale bars, 50 μm. Data are mean ± SEM from three (A and B) and two (C and D) experiments in groups of four to seven mice. *p < 0.05.

FIGURE 1.

Lack of CX3CR1 increases renal fibrosis in UUO. (AD) Fibrosis was quantified by Sirius red staining (A and B) and αSMA immunostaining (C and D) of sections of ligated or contralateral kidneys from CX3CR1-competent (Cx3cr1+/+,Cx3cr1GFP/+ [left images or white and light gray bars]) and CX3CR1-deficient (Cx3cr1GFP/GFP [right images or dark gray bars]) mice 7 d after ureter ligation. (A and C) Scale bars, 50 μm. Data are mean ± SEM from three (A and B) and two (C and D) experiments in groups of four to seven mice. *p < 0.05.

Close modal

To define the mechanisms underpinning the protective role of CX3CR1, we focused on the cells that express CX3CR1 in the kidney, namely mononuclear phagocytes, including DCs and macrophages. As reported before (3), CX3CR1-deficient (Cx3cr1GFP/GFP) mice normally possess far less GFP+ renal phagocytes than CX3CR1-competent (Cx3cr1GFP/+) mice (Fig. 2A, 2B). Also, on day 7, after ureter ligation, fewer phagocytes were seen, albeit only 10% less (Fig. 2A, 2B), suggesting the appearance of new phagocytes that partially compensated for their deficiency under homeostatic conditions. Of note, the message for the Th1 mediator Stat1 was below detection limit, and the message for the inflammatory cytokines IL-1α, IL-6, IFN-γ, and TNF and of the proresolving mediators, IL-10 and Hmox, did not differ between ligated kidneys of CX3CR1-competent and -deficient mice (Fig. 2C). Thus, the aggravated fibrosis in CX3CR1-deficient mice cannot be explained merely by stronger renal inflammation.

FIGURE 2.

Lack of CX3CR1 decreases inflammation and the abundance of renal mononuclear phagocytes in UUO. (A) Sections from contralateral Cx3cr1GFP/+ (left images) and ligated Cx3cr1GFP/GFP (right images) mice 7 d after ureteric ligation. The white bars indicate 100 μm. (B) The graph shows the numbers of GFP+ cells in Cx3cr1GFP/+ (open squares) and Cx3cr1GFP/GFP (gray circles) mice determined by counting GFP+ cells per high power field (hpf) (one representative hpf is depicted in (A)). (C) Ligated kidneys from Cx3cr1GFP/+ (white squares) or Cx3cr1GFP/GFP (gray circles) mice were homogenized, and levels of the cytokines IL-1α (top left), IL-6 (top right), (IFN-γ (bottom left), and TNF (bottom right) were determined 7 d after ureteric ligation. (D) Ligated kidneys from Cx3cr1GFP/+ (white squares) or Cx3cr1GFP/GFP (gray circles) mice were analyzed 7 d after ureteric ligation for relative Hmox RNA expression and of IL-10 protein expression. Data in (B) to (D) are mean ± SEM from two experiments in groups of four to seven mice. *p < 0.05, **p < 0.05, ***p < 0.001.

FIGURE 2.

Lack of CX3CR1 decreases inflammation and the abundance of renal mononuclear phagocytes in UUO. (A) Sections from contralateral Cx3cr1GFP/+ (left images) and ligated Cx3cr1GFP/GFP (right images) mice 7 d after ureteric ligation. The white bars indicate 100 μm. (B) The graph shows the numbers of GFP+ cells in Cx3cr1GFP/+ (open squares) and Cx3cr1GFP/GFP (gray circles) mice determined by counting GFP+ cells per high power field (hpf) (one representative hpf is depicted in (A)). (C) Ligated kidneys from Cx3cr1GFP/+ (white squares) or Cx3cr1GFP/GFP (gray circles) mice were homogenized, and levels of the cytokines IL-1α (top left), IL-6 (top right), (IFN-γ (bottom left), and TNF (bottom right) were determined 7 d after ureteric ligation. (D) Ligated kidneys from Cx3cr1GFP/+ (white squares) or Cx3cr1GFP/GFP (gray circles) mice were analyzed 7 d after ureteric ligation for relative Hmox RNA expression and of IL-10 protein expression. Data in (B) to (D) are mean ± SEM from two experiments in groups of four to seven mice. *p < 0.05, **p < 0.05, ***p < 0.001.

Close modal

These findings led us to examine in more detail the renal GFP+ phagocytes in UUO using multiphoton microscopy, which allows analyzing their morphology and motility over time. In both healthy CX3CR1-competent and CX3CR1-deficient mice, GFP+ cells with dendritic protrusions were present within the interstitium (see Supplemental Videos 1, 2).

After UUO, GFP+ cells tended to form clusters around damaged tubules (see Supplemental Videos 3, 4). In addition to GFP+ cells with dendritic morphology, the kidneys of CX3CR1-deficient mice with UUO contained larger cells (Fig. 3A) devoid of dendrites (Fig. 3B), which were more motile (Fig. 3C). Furthermore, the phagocytes in CX3CR1-deficient mice contained a subset that underwent little shape change over the time of analysis (Fig. 3D). Although these data did not reach statistical significance, they prompted us to hypothesize that GFP+ phagocytes with monocyte/macrophage properties might be involved in the increased fibrosis in CX3CR1-deficient mice with UUO.

FIGURE 3.

In vivo multiphoton microscopy of Cx3cr1GFP/+ and Cx3cr1GFP/GFP mice. (A) Analysis shows the proportion of cells with a diameter larger than 8 μm (Cx3cr1GFP/+ [open squares]; Cx3cr1GFP/GFP [gray circles]). (B and C) In the absence of CX3CR1, there is a trend (p = 0.09) toward a higher proportion of GFP+ cells without protrusions (B) and higher number of motile cells (C) (Cx3cr1GFP/+ [open squares]; Cx3cr1GFP/GFP [gray circles]). (D) Change in morphology index (ΔMI). Analysis of cellular shape change over time shows that CX3CR1-deficient mice tend to have more cells that change shape < 30 min. The x-axis in (D) represents groups of cells that are changing their shape more over time (Cx3cr1GFP/+ [open squares]; Cx3cr1GFP/GFP [gray circles]). Videos can be seen in the Supplemental Videos 1–4; data (A–D) represent the mean ± SEM of five to six mice per group.

FIGURE 3.

In vivo multiphoton microscopy of Cx3cr1GFP/+ and Cx3cr1GFP/GFP mice. (A) Analysis shows the proportion of cells with a diameter larger than 8 μm (Cx3cr1GFP/+ [open squares]; Cx3cr1GFP/GFP [gray circles]). (B and C) In the absence of CX3CR1, there is a trend (p = 0.09) toward a higher proportion of GFP+ cells without protrusions (B) and higher number of motile cells (C) (Cx3cr1GFP/+ [open squares]; Cx3cr1GFP/GFP [gray circles]). (D) Change in morphology index (ΔMI). Analysis of cellular shape change over time shows that CX3CR1-deficient mice tend to have more cells that change shape < 30 min. The x-axis in (D) represents groups of cells that are changing their shape more over time (Cx3cr1GFP/+ [open squares]; Cx3cr1GFP/GFP [gray circles]). Videos can be seen in the Supplemental Videos 1–4; data (A–D) represent the mean ± SEM of five to six mice per group.

Close modal

We hypothesized that higher numbers of renal macrophages might explain the profibrotic effect of CX3CR1 deficiency in UUO (Fig. 1) because it has been previously reported that renal macrophages but not DCs mediate fibrosis in this model (16, 18, 66). However, Cx3cr1GFP/GFP mice possessed similar or lower numbers of GFP+ renal phagocytes under homeostatic conditions, in experimental glomerulonephritis and in kidney infection compared with CX3CR1-competent controls (3, 58), consistent with our present findings in UUO (Fig. 2A, 2B). We therefore discriminated between GFP+ kidney macrophages and DCs by costaining with F4/80 and CD11c. We found that CX3CR1 deficiency reduced renal CD11c+ DC numbers in the ligated and contralateral kidney (Fig. 4A, upper gate in the dot plots) consistent with the morphological changes shown in Fig. 3. Next, we focused on CD11cGFP+ cells (Fig. 4A, lower gate in the dot plots). We found that >90% of these cells expressed F4/80 (Fig. 4B) identifying these GFP/CX3CR1-expressing cells as macrophages. Furthermore, these data also exclude considerable contaminations of other cells than monocytes/macrophages and DCs in the microscopy data (Figs. 2, 3). Because the microscopy findings above (Fig. 3) hinted at the appearance of monocytes/macrophages, we focused on the analysis of macrophages expressing the Gr1 marker, which designates mononuclear phagocytes recently recruited from the circulation (67, 68) and which is related to helper macrophages expressing Ly6C (4), a component of Gr1. Gr1+ macrophages were considerably more numerous already on day 3 after UUO (Fig. 4B, 4C). In the absence of CX3CR1, these cells were more abundant than in CX3CR1-competent mice, whereas the abundance of kidney-resident Gr1 macrophages was unchanged (Fig. 4C). On day 7, fewer recruited macrophages were seen, whereas numbers of Gr1 macrophages had further increased (Fig. 4C). Thus, the lack of CX3CR1 reduced the number of CD11c+ DCs, but increased the numbers of CD11c Gr1+ macrophages in UUO. This shift from DCs to macrophages explains why the total number of GFP+ phagocytes was only somewhat reduced in CX3CR1-deficient mice (Fig. 2), and the relative increase of macrophages might explain the increased kidney fibrosis in the mice.

FIGURE 4.

CX3CR1 differentially regulates the numbers of renal DCs and Gr1+ macrophages and TGF-β production in UUO. (A) Flow cytometric analysis of single-cell suspensions from nonligated (healthy; left), contralateral (middle), and ligated kidneys (right) of Cx3cr1GFP/+ (upper row) and Cx3cr1GFP/GFP (lower row) mice. Shown is CD11c staining for DCs versus CX3CR1 expression levels (GFP) on CD45+ immune cells. Note that homozygous Cx3cr1GFP/GFP mice show two times stronger GFP signals. (B) The CD11cGFP+ macrophages [bottom right region in (A)] were gated and their F4/80 versus Gr1 expression was displayed to identify Gr1 and Gr1+ macrophages (top right and left region). (C) Enumeration of CD11cCD45+GFP+F4/80+ macrophages (Gr1 macrophages in the left panel, Gr1+ macrophages in the middle panel) and CD11c+ CD45+ DCs (right panel) in healthy mice (d0) and on days 3 and 7 after UUO. (D) Three days after ureteric ligation, intracellular content of the profibrotic factor TGF-β was determined on F4/80+GFP+CD11c macrophages in ligated and contralateral kidneys as indicated and the numbers of these TGFβ+F4/80+GFP+CD11c macrophages expressing Gr1 or not were displayed. Dot plots in (A) represent concatenated dot plots from four to seven mice. Data represent the mean ± SEM from three (A–C) and four (D) individual experiments in groups of four to seven mice. *p < 0.05, **p < 0.01, ***p < 0.001.

FIGURE 4.

CX3CR1 differentially regulates the numbers of renal DCs and Gr1+ macrophages and TGF-β production in UUO. (A) Flow cytometric analysis of single-cell suspensions from nonligated (healthy; left), contralateral (middle), and ligated kidneys (right) of Cx3cr1GFP/+ (upper row) and Cx3cr1GFP/GFP (lower row) mice. Shown is CD11c staining for DCs versus CX3CR1 expression levels (GFP) on CD45+ immune cells. Note that homozygous Cx3cr1GFP/GFP mice show two times stronger GFP signals. (B) The CD11cGFP+ macrophages [bottom right region in (A)] were gated and their F4/80 versus Gr1 expression was displayed to identify Gr1 and Gr1+ macrophages (top right and left region). (C) Enumeration of CD11cCD45+GFP+F4/80+ macrophages (Gr1 macrophages in the left panel, Gr1+ macrophages in the middle panel) and CD11c+ CD45+ DCs (right panel) in healthy mice (d0) and on days 3 and 7 after UUO. (D) Three days after ureteric ligation, intracellular content of the profibrotic factor TGF-β was determined on F4/80+GFP+CD11c macrophages in ligated and contralateral kidneys as indicated and the numbers of these TGFβ+F4/80+GFP+CD11c macrophages expressing Gr1 or not were displayed. Dot plots in (A) represent concatenated dot plots from four to seven mice. Data represent the mean ± SEM from three (A–C) and four (D) individual experiments in groups of four to seven mice. *p < 0.05, **p < 0.01, ***p < 0.001.

Close modal

Next, we examined whether CX3CR1 also modulates the functionality of macrophages. To this end, we analyzed the production of TGF-β, the key profibrotic molecule in the kidney (65, 69). Most of the TGF-β–producing leukocytes indeed expressed F4/80 (Supplemental Fig. 1). Importantly, the frequencies (Supplemental Fig. 1) and numbers (Fig. 4D) of TGF-β–producing Gr1 and Gr1+ macrophages were increased in ligated kidneys of CX3CR1-deficient mice. M1/M2 differentiation markers such as Arg1, Mrc1, Irf5, Stat6, and Retnla1 were little altered in CX3CR1 deficiency (Fig. 5), indicating that the functional changes of macrophages could not be explained by effects on M1/M2 differentiation (70, 71). These data demonstrate that CX3CR1 not only negatively regulates the numbers of profibrotic macrophages but also their fibrogenic potential.

FIGURE 5.

Polarization of macrophages does not depend on CX3CR1. Gene expression profiles in contralateral and ligated kidneys 3 d after ureteric ligation. Data are normalized to the housekeeping gene Hprt and represent the mean ± SEM from two individual experiments in groups of four to six mice.

FIGURE 5.

Polarization of macrophages does not depend on CX3CR1. Gene expression profiles in contralateral and ligated kidneys 3 d after ureteric ligation. Data are normalized to the housekeeping gene Hprt and represent the mean ± SEM from two individual experiments in groups of four to six mice.

Close modal

The inhibitory effects of CX3CR1 on renal macrophages could theoretically be due to reduced recruitment of macrophage precursors such as monocytes, reduced intrarenal differentiation into macrophages, reduced proliferation, increased survival or be due to a combination of these factors. Therefore, we examined all of these potential mechanisms.

To establish a system to analyze macrophage precursor recruitment to the kidney, we first adoptively transferred bone marrow–derived monocytes expressing a YFP reporter for CD11c, which allows convenient discrimination between DCs and macrophages, into CD45.1-congenic mice with or without UUO. Only a few monocytes reached the kidneys of healthy mice and the contralateral kidney of mice with UUO (Fig. 6A), suggesting that renal phagocytes possess a low turnover rate. In contrast, ligated kidneys contained clearly detectable monocytes 2 d after transfer, and some of them were YFP+ (Fig. 6A), indicating differentiation into DCs.

FIGURE 6.

Lack of CX3CR1 decreases phagocyte recruitment into the kidney. (A) Sorted CD19CD3CD11c bone marrow cells from CD45.2 CD11cYFP mice were adoptively transferred into CD45.1 wild-type mice. The frequency of donor YFP+CD45.2+F4/80+CD11c+ DCs (upper region) and of YFPCD45.2+F4/80+CD11c macrophages (lower region) in the kidney was determined 3 d after ureter ligation (bone marrow cells were transferred 1 d after ligation). (B) Sorted CD19CD3CD11c bone marrow cells from Cx3cr1GFP/+ mice were transferred into wild-type mice 1 d after ligation. Three days after ureter ligation GFP+F4/80+CD11c+ DCs (upper region) and GFP+F4/80+CD11c macrophages (lower region) were detected by flow cytometry. (C) Sorted CD19CD3CD11c bone marrow cells from Cx3cr1GFP/+ or Cx3cr1GFP/GFP mice were transferred into wild-type mice. Three days after ureter ligation, the number of F4/80+GFP+CD11c macrophages (left panel) and of GFP+F4/80+CD11c+ DCs (right panel) were determined by flow cytometry. The group designated without transfer was used to control for specific detection of transferred cells. (D) Kidneys from Cx3cr1GFP/+ (white squares) or Cx3cr1GFP/GFP (gray circles) mice were homogenized and levels of the chemokines MCP-1 (top left), MCP-3 (top right), MIP1a (bottom left), and RANTES (bottom right) were determined 3 d after ureter ligation. Control animals did not underwent unilateral ureteral ligation. Data represent three (A–C) and two (D) experiments in groups of five to six mice. Results in (D) are mean ± SEM. *p < 0.05, **p < 0.01.

FIGURE 6.

Lack of CX3CR1 decreases phagocyte recruitment into the kidney. (A) Sorted CD19CD3CD11c bone marrow cells from CD45.2 CD11cYFP mice were adoptively transferred into CD45.1 wild-type mice. The frequency of donor YFP+CD45.2+F4/80+CD11c+ DCs (upper region) and of YFPCD45.2+F4/80+CD11c macrophages (lower region) in the kidney was determined 3 d after ureter ligation (bone marrow cells were transferred 1 d after ligation). (B) Sorted CD19CD3CD11c bone marrow cells from Cx3cr1GFP/+ mice were transferred into wild-type mice 1 d after ligation. Three days after ureter ligation GFP+F4/80+CD11c+ DCs (upper region) and GFP+F4/80+CD11c macrophages (lower region) were detected by flow cytometry. (C) Sorted CD19CD3CD11c bone marrow cells from Cx3cr1GFP/+ or Cx3cr1GFP/GFP mice were transferred into wild-type mice. Three days after ureter ligation, the number of F4/80+GFP+CD11c macrophages (left panel) and of GFP+F4/80+CD11c+ DCs (right panel) were determined by flow cytometry. The group designated without transfer was used to control for specific detection of transferred cells. (D) Kidneys from Cx3cr1GFP/+ (white squares) or Cx3cr1GFP/GFP (gray circles) mice were homogenized and levels of the chemokines MCP-1 (top left), MCP-3 (top right), MIP1a (bottom left), and RANTES (bottom right) were determined 3 d after ureter ligation. Control animals did not underwent unilateral ureteral ligation. Data represent three (A–C) and two (D) experiments in groups of five to six mice. Results in (D) are mean ± SEM. *p < 0.05, **p < 0.01.

Close modal

To investigate whether CX3CR1 is required for the entry of these precursors into the kidney, we transferred monocytes from Cx3cr1GFP/GFP mice or Cx3cr1GFP/+ mice into CX3CR1-competent wild-type recipients 1 d after ureter ligation (Fig. 6B, 6C). After an additional 2 d, less CX3CR1-deficient phagocytes had entered the ligated kidneys than did CX3CR1-competent phagocytes (Fig. 6C). Notably, no difference was seen in the blood (Supplemental Fig. 2), excluding an effect of CX3CR1 on the abundance of circulating macrophage precursors. Thus, CX3CR1 promoted, rather than inhibited, phagocyte precursor recruitment. Furthermore, the levels of the chemokine MIP-1α that can attract macrophage precursors were decreased in ligated and also in contralateral kidneys of CX3CR1-deficient mice, and there were trends toward reduced levels of RANTES, MCP-1, and MCP-3 (Fig. 6D). This is consistent with reduced macrophage precursor recruitment and may be explained by the reduction in chemokine-producing DCs in CX3CR1-deficient mice (3).

To study whether CX3CR1 regulated the differentiation of monocytes into macrophages versus DCs, we determined the ratio between donor-derived DCs and macrophages after entering the ligated kidney. This parameter was not significantly changed in CX3CR1 deficiency (Fig. 7A). Furthermore, the intrarenal levels of cytokines that affect DC/macrophage differentiation, GM-CSF or M-CSF, were CX3CR1 independent (Fig. 7B). Taken together, these findings argue against changes in macrophage recruitment or differentiation as an explanation for their increased intrarenal numbers in CX3CR1-deficient mice.

FIGURE 7.

Differentiation of infiltrating monocytes into DCs is CX3CR1-independent. (A) Sorted CD19CD3CD11c bone marrow cells from Cx3cr1GFP/+ or Cx3cr1GFP/GFP mice were transferred into wild-type mice. Three days after ureter ligation, the number of F4/80+GFP+CD11c macrophages and of GFP+F4/80+CD11c+ DCs were determined by flow cytometry, and the ratio between these DCs and macrophages were calculated (number of DCs per macrophage). (B) Levels of GM-CSF (left) and M-CSF (right) in homogenates of ligated kidneys from Cx3cr1GFP/+ (white squares) and Cx3cr1GFP/GFP (gray circles) mice 3 d after UUO in comparison with contralateral kidneys. Results are mean ± SEM and represent three (A) and two (B) independent experiments in groups of 5–10 mice.

FIGURE 7.

Differentiation of infiltrating monocytes into DCs is CX3CR1-independent. (A) Sorted CD19CD3CD11c bone marrow cells from Cx3cr1GFP/+ or Cx3cr1GFP/GFP mice were transferred into wild-type mice. Three days after ureter ligation, the number of F4/80+GFP+CD11c macrophages and of GFP+F4/80+CD11c+ DCs were determined by flow cytometry, and the ratio between these DCs and macrophages were calculated (number of DCs per macrophage). (B) Levels of GM-CSF (left) and M-CSF (right) in homogenates of ligated kidneys from Cx3cr1GFP/+ (white squares) and Cx3cr1GFP/GFP (gray circles) mice 3 d after UUO in comparison with contralateral kidneys. Results are mean ± SEM and represent three (A) and two (B) independent experiments in groups of 5–10 mice.

Close modal

We next investigated whether CX3CR1 regulates apoptosis by staining with Annexin V. We found that more Gr1 and Gr1+ macrophages were apoptotic within the ligated kidneys (Fig. 8A, 8B, black line versus gray line). However, lack of CX3CR1 did not reduce but instead increased the frequency of Annexin V+Gr1 and Gr1+ macrophages (Fig. 8A, 8B), excluding apoptosis as the mechanism for the increased number of macrophages in CX3Cr1-deficient animals.

FIGURE 8.

Lack of CX3CR1 increases apoptosis of renal macrophages in UUO. (A and B) Gr1 (A) and Gr1+ (B) macrophages (CD45+CD11cGFP+F4/80+; gating strategy, see Fig. 4A, 4B) from ligated (black thick line) and contralateral (gray line) kidneys from Cx3cr1GFP/+ (left panels) and Cx3cr1GFP/GFP (right panels) mice were analyzed by flow cytometry for annexin V binding on day 3 after ureter ligation. The gray area shows fluorescence without addition of annexin V. Data are mean ± SEM and the graphs represent concatenated histograms from seven to eight mice (concatenated = combined histograms from seven to eight mice).

FIGURE 8.

Lack of CX3CR1 increases apoptosis of renal macrophages in UUO. (A and B) Gr1 (A) and Gr1+ (B) macrophages (CD45+CD11cGFP+F4/80+; gating strategy, see Fig. 4A, 4B) from ligated (black thick line) and contralateral (gray line) kidneys from Cx3cr1GFP/+ (left panels) and Cx3cr1GFP/GFP (right panels) mice were analyzed by flow cytometry for annexin V binding on day 3 after ureter ligation. The gray area shows fluorescence without addition of annexin V. Data are mean ± SEM and the graphs represent concatenated histograms from seven to eight mice (concatenated = combined histograms from seven to eight mice).

Close modal

It has been previously reported that tissue macrophage numbers can be regulated by their local proliferation (72, 73). To investigate whether CX3CR1 decreases intrarenal macrophage numbers by inhibiting their proliferation, we determined expression of the proliferation marker Ki67 on day 3 and uptake of BrdU from day 3 until day 5 after ureteric ligation. We found that Ki67 expression was significantly increased in macrophages of ligated kidneys from CX3CR1-deficient mice compared with CX3CR1-competent mice (Fig. 9A), and this was seen in both Gr1 and Gr1+ macrophages (Fig. 9B). Furthermore, more macrophages of CX3CR1-deficient mice incorporated BrdU (Fig. 9C, 9D), and the uptake per macrophage was higher (Fig. 9E), verifying local CX3CR1-dependent proliferation of macrophages. Notably, such increased proliferation was not detectable in DCs (Fig. 9D). Thus, CX3CR1 inhibits local macrophage proliferation in UUO, and this can explain the higher numbers of profibrotic renal macrophages in the absence of CX3CR1.

FIGURE 9.

Lack of CX3CR1 increases proliferation of Gr1 and of Gr1+ macrophages in UUO. (A) Three days after ureter ligation, intracellular expression of the proliferation marker Ki67 was determined on F4/80+GFP+CD11c macrophages in Cx3cr1GFP/+ (middle dot plot) and Cx3cr1GFP/GFP (right dot plot) mice. Isotype Ab staining shows the negative control to ensure Ki67 specificity (left dot plot). (B) Percentages of Ki67+F4/80+GFP+CD11c macrophages expressing Gr1 (right) or not (left) in the ligated or contralateral kidneys of Cx3cr1GFP/+ (white squares) and Cx3cr1GFP/GFP (gray circles) mice on day 3 after ureter ligation. (C) BrdU was injected into Cx3cr1GFP/+ and Cx3cr1GFP/GFP mice 3 and 4 d after UUO. One day later (day 5), F4/80+GFP+CD11c macrophages in the ligated kidneys were analyzed for their BrdU incorporation. (D) Percentages of BrdU+F4/80+GFP+CD11c macrophages and BrdU+CD11c+ DCs of Cx3cr1GFP/+ (white squares) and Cx3cr1GFP/GFP (gray circles) mice on day 5 after ureter ligation (BrdU was administered on day 3 and 4 after ligation). (E) BrdU was injected into Cx3cr1GFP/+ and Cx3cr1GFP/GFP mice 3 and 4 d after UUO. One day later (day 5), F4/80+GFP+CD11c macrophages in the ligated kidneys were analyzed for their BrdU incorporation. Concatenated dot plots in (A) are combined data from six mice. Data (B–D) represent the mean ± SEM in groups of four to six mice from three experiments. *p < 0.05, **p < 0.01.

FIGURE 9.

Lack of CX3CR1 increases proliferation of Gr1 and of Gr1+ macrophages in UUO. (A) Three days after ureter ligation, intracellular expression of the proliferation marker Ki67 was determined on F4/80+GFP+CD11c macrophages in Cx3cr1GFP/+ (middle dot plot) and Cx3cr1GFP/GFP (right dot plot) mice. Isotype Ab staining shows the negative control to ensure Ki67 specificity (left dot plot). (B) Percentages of Ki67+F4/80+GFP+CD11c macrophages expressing Gr1 (right) or not (left) in the ligated or contralateral kidneys of Cx3cr1GFP/+ (white squares) and Cx3cr1GFP/GFP (gray circles) mice on day 3 after ureter ligation. (C) BrdU was injected into Cx3cr1GFP/+ and Cx3cr1GFP/GFP mice 3 and 4 d after UUO. One day later (day 5), F4/80+GFP+CD11c macrophages in the ligated kidneys were analyzed for their BrdU incorporation. (D) Percentages of BrdU+F4/80+GFP+CD11c macrophages and BrdU+CD11c+ DCs of Cx3cr1GFP/+ (white squares) and Cx3cr1GFP/GFP (gray circles) mice on day 5 after ureter ligation (BrdU was administered on day 3 and 4 after ligation). (E) BrdU was injected into Cx3cr1GFP/+ and Cx3cr1GFP/GFP mice 3 and 4 d after UUO. One day later (day 5), F4/80+GFP+CD11c macrophages in the ligated kidneys were analyzed for their BrdU incorporation. Concatenated dot plots in (A) are combined data from six mice. Data (B–D) represent the mean ± SEM in groups of four to six mice from three experiments. *p < 0.05, **p < 0.01.

Close modal

The chemokine receptor CX3CR1 is widely expressed by DCs and macrophages (74), but its functional role is still not fully clear. In this paper, we report a previously undescribed antifibrotic effect of CX3CR1: it reduced the inflammation-induced proliferation of TGF-β–producing renal macrophages, which are critically important in experimental kidney fibrosis (1518). This finding was unexpected, given the proinflammatory and profibrotic properties of CX3CR1 not only in diseases of the kidney (3, 49, 54, 5759, 65), but also of the lung (75), liver (50), intestine (28), skin (55), arteriosclerotic vessels (76), and the CNS (33). CX3CR1 has been shown to contribute to the inflammatory recruitment of monocytes from the circulation into all these organs (44). We found that CX3CR1 had a similar effect in UUO so that fewer monocytes lacking this receptor entered the inflamed kidneys. The lower levels of monocyte-attracting chemokines and of inflammatory mediators that we detected in kidneys of CX3CR1-deficient mice may be responsible for the reduced kidney recruitment of these cells. It is likely that these cells originated from monocytes, which have been shown to give rise to tissue macrophages under inflammatory conditions (5). The reduction of monocyte-attracting chemokines likely resulted from the paucity of DCs in kidneys of CX3CR1-deficient animals because DCs are the principal producers of chemokines and inflammatory mediators in the kidney (7779). However, these findings were hard to reconcile with the accumulation of macrophages in CX3CR1-deficient mice, implying that other mechanisms must exist that overcompensated for the reduced attraction of macrophage precursors.

One such mechanism might be a negative effect of CX3CR1 on the survival of tissue macrophages. However, previous studies in disease models of liver, CNS, gut, and arteriosclerotic vessels (74) demonstrated that CX3CR1 signaling increased rather than reduced the life span of macrophages. In contrast to these findings, one recent study reported increased macrophage numbers and liver fibrosis in CX3CR1-deficient mice after surgical bile duct ligation, a model that resembles UUO by being induced by mechanic tissue damage (50). The discrepancy between macrophage accumulation and decreased macrophage life span in that study was suggested to have resulted from the release of proinflammatory mediators from CX3CR1-deficient dying macrophages that attracted further profibrotic macrophages. Although we did observe more dying macrophages after UUO in CX3CR1-deficient mice, we found neither increased monocyte-attracting chemokines and inflammatory cytokines, nor did we find a stronger influx of inflammatory monocytes, arguing against inflammation resulting from premature macrophage death in the kidney as the underlying reason for macrophage accumulation.

Another hypothetic mechanism is that CX3CR1 might cause immigrating monocytes to differentiate preferentially into macrophages rather than DCs. DC numbers are severely reduced in CX3CR1-deficient mice (3), which would be consistent with altered differentiation. DCs do not play a major role in UUO, whereas macrophages are critical (18, 49, 54). This is not unexpected given that DCs primarily regulate adaptive immune responses, explaining why they affected fibrosis in models that involve adaptive immune cells, such as ischemia/reperfusion injury, crescentic glomerulonephritis and hypertensive nephropathy (3, 49, 54, 57). By contrast, a model of mechanic tissue injury like UUO is more likely mediated by cells of the innate immune system, like macrophages. However, adoptive transfer experiments revealed that monocytes expressing or lacking CX3CR1 gave rise to similar ratios between macrophages and DCs, arguing against an effect of CX3CR1-signaling on the monocytes' decision to differentiate into either DCs or macrophages. This conclusion was corroborated by unchanged levels of the growth factors that usually govern this decision, M-CSF and GM-CSF.

In vivo multiphoton microscopic analysis revealed morphologic alterations of CX3CR1-deficient renal phagocytes. Although there were slight changes in the morphology of CX3CR1+ kidney DCs, such as more yet shorter dendrites (our unpublished observations), it is unlikely that these alterations impacted fibrogenesis given that DCs do not affect renal fibrosis (18, 49, 54). However, in addition to resident DCs, two-photon microscopy revealed larger and rounder CX3CR1+ cells that were more motile and maintained their shape over time, which is consistent with the morphology and dynamics of monocytes/macrophages. These cells were more abundant in CX3CR1-deficient mice, prompting us to hypothesize about a functional link between macrophages and CX3CR1. Indeed, we noted an inhibitory effect of CX3CR1 on intrarenal macrophage proliferation. This can explain why these cells accumulated in CX3CR1-deficient mice, although their precursor recruitment was less efficient and although more of them were apoptotic. As we excluded other potential causes, increased proliferation was most likely the cause for the accumulation of macrophages in the CX3CR1-deficient kidney. It is possible that CX3CR1 also prevents macrophage proliferation in DC-dependent models of renal disease (e.g., crescentic glomerulonephritis) but that this anti-inflammatory effect does not become manifest because its proinflammatory effect on the more numerous DCs prevails.

Traditionally, tissue macrophages were thought to arise from circulating monocytes recruited in response to locally produced chemokines (11). Recent findings have revealed that some tissue macrophages are derived from embryonic precursors (22). A sudden demand because of inflammatory conditions is met by the proliferation of such tissue-resident macrophages, in particular under Th2 or M2-associated conditions (72, 73, 80). We found that all macrophages, including those that had just recently been recruited to the kidney, showed stronger proliferation in the absence of CX3CR1. These findings are consistent with the concept that local proliferation of tissue macrophages regulates their numbers and extend this concept by showing that also recently recruited macrophages are locally regulated in the inflamed kidney. It remains to be seen whether this mechanism also contributes to fibrosis in other organs.

CX3CR1 not only affected the abundance, but also the functional state of renal macrophages. In its absence, more macrophages produced TGF-β, the key profibrotic mediator in the kidney (69). A previous study showed that CCR2-dependent macrophages produced TGF-β in UUO and that the lack of these cells significantly reduced renal fibrosis (16). Also, we found that both Gr1 and Gr1+ macrophages produced TGF-β and that production was increased in the absence of CX3CR1. TGFβ production suggested acquisition of a profibrotic functional state change. This state could not be explained by M2 differentiation, nor was it associated with higher renal inflammation. Recently, the classical M1/M2 paradigm was refined by the discovery of an activation-independent functional state that does not fall into the M1 or M2 definition (71). Our findings are consistent with this notion by demonstrating that TGF-β–producing macrophages cannot be categorized as classical M1 and M2 macrophages. Future studies are required to clarify whether these profibrotic macrophages belong to a distinct functional state.

In summary, we report a novel inhibitory effect of CX3CR1 on local macrophage proliferation and on fibrogenesis in the kidney. In crescentic GN, the previously described proinflammatory influence of CX3CR1 on DCs prevailed (3) because macrophages are downstream of DCs in the pathomechanism of this disease and because DCs outnumber macrophages in the kidney. However, in DC-independent renal diseases like UUO, the inhibitory effect of CX3CR1 on macrophages becomes manifest, as we demonstrated in the present study, so that CX3CR1 inhibition in such diseases aggravates fibrosis. This may apply also to organs in which CX3CR1 does not regulate DCs, like the liver or the lung (3). In this case, therapeutic CX3CR1 inhibition in liver and lung disease may turn out profibrotic.

We thank F. Tacke for critically reading the manuscript and Dr. Camden Lo for assistance with in vivo imaging analytic parameters. We acknowledge support by the Flow Cytometry Core Facility and the House for Experimental Therapy of the medical faculty of Bonn University.

This work was supported by the Deutsche Forschungsgemeinschaft (Grants SFB Transregio 57, Klinische Forschergruppe 228, and BO 3755/1-1) and the Gottfried-Wilhelm Leibniz Award (to C.K.) and the Australian National Health and Medical Research Council (Project Grants 565033 and 1045065). C.K. and D.R.E. are members of the Deutsche Forschungsgemeinschaft Excellence Cluster ImmunoSensation.

The online version of this article contains supplemental material.

Abbreviations used in this article:

DC

dendritic cell

ΔMI

change in morphology index

αSMA

α-smooth muscle actin

UUO

unilateral ureter ligation.

1
Nelson
P. J.
,
Rees
A. J.
,
Griffin
M. D.
,
Hughes
J.
,
Kurts
C.
,
Duffield
J.
.
2012
.
The renal mononuclear phagocytic system.
J. Am. Soc. Nephrol.
23
:
194
203
.
2
Kurts
C.
,
Panzer
U.
,
Anders
H. J.
,
Rees
A. J.
.
2013
.
The immune system and kidney disease: basic concepts and clinical implications.
Nat. Rev. Immunol.
13
:
738
753
.
3
Hochheiser
K.
,
Heuser
C.
,
Krause
T. A.
,
Teteris
S.
,
Ilias
A.
,
Weisheit
C.
,
Hoss
F.
,
Tittel
A. P.
,
Knolle
P. A.
,
Panzer
U.
, et al
.
2013
.
Exclusive CX3CR1 dependence of kidney DCs impacts glomerulonephritis progression.
J. Clin. Invest.
123
:
4242
4254
.
4
Schiwon
M.
,
Weisheit
C.
,
Franken
L.
,
Gutweiler
S.
,
Dixit
A.
,
Meyer-Schwesinger
C.
,
Pohl
J. M.
,
Maurice
N. J.
,
Thiebes
S.
,
Lorenz
K.
, et al
.
2014
.
Crosstalk between sentinel and helper macrophages permits neutrophil migration into infected uroepithelium.
Cell
156
:
456
468
.
5
Geissmann
F.
,
Gordon
S.
,
Hume
D. A.
,
Mowat
A. M.
,
Randolph
G. J.
.
2010
.
Unravelling mononuclear phagocyte heterogeneity.
Nat. Rev. Immunol.
10
:
453
460
.
6
LeibundGut-Landmann
S.
,
Gross
O.
,
Robinson
M. J.
,
Osorio
F.
,
Slack
E. C.
,
Tsoni
S. V.
,
Schweighoffer
E.
,
Tybulewicz
V.
,
Brown
G. D.
,
Ruland
J.
,
Reis e Sousa
C.
.
2007
.
Syk- and CARD9-dependent coupling of innate immunity to the induction of T helper cells that produce interleukin 17.
Nat. Immunol.
8
:
630
638
.
7
Krüger
T.
,
Benke
D.
,
Eitner
F.
,
Lang
A.
,
Wirtz
M.
,
Hamilton-Williams
E. E.
,
Engel
D.
,
Giese
B.
,
Müller-Newen
G.
,
Floege
J.
,
Kurts
C.
.
2004
.
Identification and functional characterization of dendritic cells in the healthy murine kidney and in experimental glomerulonephritis.
J. Am. Soc. Nephrol.
15
:
613
621
.
8
Miller
J. C.
,
Brown
B. D.
,
Shay
T.
,
Gautier
E. L.
,
Jojic
V.
,
Cohain
A.
,
Pandey
G.
,
Leboeuf
M.
,
Elpek
K. G.
,
Helft
J.
, et al
.
2012
.
Deciphering the transcriptional network of the dendritic cell lineage.
Nat. Immunol.
13
:
888
899
.
9
Gautier
E. L.
,
Shay
T.
,
Miller
J.
,
Greter
M.
,
Jakubzick
C.
,
Ivanov
S.
,
Helft
J.
,
Chow
A.
,
Elpek
K. G.
,
Gordonov
S.
, et al
.
2012
.
Gene-expression profiles and transcriptional regulatory pathways that underlie the identity and diversity of mouse tissue macrophages.
Nat. Immunol.
13
:
1118
1128
.
10
Tittel
A. P.
,
Heuser
C.
,
Ohliger
C.
,
Knolle
P. A.
,
Engel
D. R.
,
Kurts
C.
.
2011
.
Kidney dendritic cells induce innate immunity against bacterial pyelonephritis.
J. Am. Soc. Nephrol.
22
:
1435
1441
.
11
Gordon
S.
,
Taylor
P. R.
.
2005
.
Monocyte and macrophage heterogeneity.
Nat. Rev. Immunol.
5
:
953
964
.
12
Murray
P. J.
,
Wynn
T. A.
.
2011
.
Protective and pathogenic functions of macrophage subsets.
Nat. Rev. Immunol.
11
:
723
737
.
13
Vernon
M. A.
,
Mylonas
K. J.
,
Hughes
J.
.
2010
.
Macrophages and renal fibrosis.
Semin. Nephrol.
30
:
302
317
.
14
Wynn
T. A.
,
Barron
L.
.
2010
.
Macrophages: master regulators of inflammation and fibrosis.
Semin. Liver Dis.
30
:
245
257
.
15
Persy
V. P.
,
Verhulst
A.
,
Ysebaert
D. K.
,
De Greef
K. E.
,
De Broe
M. E.
.
2003
.
Reduced postischemic macrophage infiltration and interstitial fibrosis in osteopontin knockout mice.
Kidney Int.
63
:
543
553
.
16
Kitagawa
K.
,
Wada
T.
,
Furuichi
K.
,
Hashimoto
H.
,
Ishiwata
Y.
,
Asano
M.
,
Takeya
M.
,
Kuziel
W. A.
,
Matsushima
K.
,
Mukaida
N.
,
Yokoyama
H.
.
2004
.
Blockade of CCR2 ameliorates progressive fibrosis in kidney.
Am. J. Pathol.
165
:
237
246
.
17
Kitamoto
K.
,
Machida
Y.
,
Uchida
J.
,
Izumi
Y.
,
Shiota
M.
,
Nakao
T.
,
Iwao
H.
,
Yukimura
T.
,
Nakatani
T.
,
Miura
K.
.
2009
.
Effects of liposome clodronate on renal leukocyte populations and renal fibrosis in murine obstructive nephropathy.
J. Pharmacol. Sci.
111
:
285
292
.
18
Snelgrove
S. L.
,
Kausman
J. Y.
,
Lo
C.
,
Lo
C.
,
Ooi
J. D.
,
Coates
P. T.
,
Hickey
M. J.
,
Holdsworth
S. R.
,
Kurts
C.
,
Engel
D. R.
,
Kitching
A. R.
.
2012
.
Renal dendritic cells adopt a pro-inflammatory phenotype in obstructive uropathy to activate T cells but do not directly contribute to fibrosis.
Am. J. Pathol.
180
:
91
103
.
19
Wynn
T. A.
2008
.
Cellular and molecular mechanisms of fibrosis.
J. Pathol.
214
:
199
210
.
20
Duffield
J. S.
,
Lupher
M.
,
Thannickal
V. J.
,
Wynn
T. A.
.
2013
.
Host responses in tissue repair and fibrosis.
Annu. Rev. Pathol.
8
:
241
276
.
21
Ginhoux
F.
,
Tacke
F.
,
Angeli
V.
,
Bogunovic
M.
,
Loubeau
M.
,
Dai
X. M.
,
Stanley
E. R.
,
Randolph
G. J.
,
Merad
M.
.
2006
.
Langerhans cells arise from monocytes in vivo.
Nat. Immunol.
7
:
265
273
.
22
Schulz
C.
,
Gomez Perdiguero
E.
,
Chorro
L.
,
Szabo-Rogers
H.
,
Cagnard
N.
,
Kierdorf
K.
,
Prinz
M.
,
Wu
B.
,
Jacobsen
S. E.
,
Pollard
J. W.
, et al
.
2012
.
A lineage of myeloid cells independent of Myb and hematopoietic stem cells.
Science
336
:
86
90
.
23
Shi
C.
,
Pamer
E. G.
.
2011
.
Monocyte recruitment during infection and inflammation.
Nat. Rev. Immunol.
11
:
762
774
.
24
Ginhoux
F.
,
Greter
M.
,
Leboeuf
M.
,
Nandi
S.
,
See
P.
,
Gokhan
S.
,
Mehler
M. F.
,
Conway
S. J.
,
Ng
L. G.
,
Stanley
E. R.
, et al
.
2010
.
Fate mapping analysis reveals that adult microglia derive from primitive macrophages.
Science
330
:
841
845
.
25
Schraml
B. U.
,
van Blijswijk
J.
,
Zelenay
S.
,
Whitney
P. G.
,
Filby
A.
,
Acton
S. E.
,
Rogers
N. C.
,
Moncaut
N.
,
Carvajal
J. J.
,
Reis e Sousa
C.
.
2013
.
Genetic tracing via DNGR-1 expression history defines dendritic cells as a hematopoietic lineage.
Cell
154
:
843
858
.
26
Kezic
J.
,
McMenamin
P. G.
.
2010
.
The monocyte chemokine receptor CX3CR1 does not play a significant role in the pathogenesis of experimental autoimmune uveoretinitis.
Invest. Ophthalmol. Vis. Sci.
51
:
5121
5127
.
27
Auffray
C.
,
Fogg
D. K.
,
Narni-Mancinelli
E.
,
Senechal
B.
,
Trouillet
C.
,
Saederup
N.
,
Leemput
J.
,
Bigot
K.
,
Campisi
L.
,
Abitbol
M.
, et al
.
2009
.
CX3CR1+CD115+CD135+ common macrophage/DC precursors and the role of CX3CR1 in their response to inflammation.
J. Exp. Med.
206
:
595
606
.
28
Medina-Contreras
O.
,
Geem
D.
,
Laur
O.
,
Williams
I. R.
,
Lira
S. A.
,
Nusrat
A.
,
Parkos
C. A.
,
Denning
T. L.
.
2011
.
CX3CR1 regulates intestinal macrophage homeostasis, bacterial translocation, and colitogenic Th17 responses in mice.
J. Clin. Invest.
121
:
4787
4795
.
29
Farache
J.
,
Zigmond
E.
,
Shakhar
G.
,
Jung
S.
.
2013
.
Contributions of dendritic cells and macrophages to intestinal homeostasis and immune defense.
Immunol. Cell Biol.
91
:
232
239
.
30
Kostadinova
F. I.
,
Baba
T.
,
Ishida
Y.
,
Kondo
T.
,
Popivanova
B. K.
,
Mukaida
N.
.
2010
.
Crucial involvement of the CX3CR1-CX3CL1 axis in dextran sulfate sodium-mediated acute colitis in mice.
J. Leukoc. Biol.
88
:
133
143
.
31
Landsman
L.
,
Varol
C.
,
Jung
S.
.
2007
.
Distinct differentiation potential of blood monocyte subsets in the lung.
J. Immunol.
178
:
2000
2007
.
32
Jakubzick
C.
,
Tacke
F.
,
Ginhoux
F.
,
Wagers
A. J.
,
van Rooijen
N.
,
Mack
M.
,
Merad
M.
,
Randolph
G. J.
.
2008
.
Blood monocyte subsets differentially give rise to CD103+ and CD103 pulmonary dendritic cell populations.
J. Immunol.
180
:
3019
3027
.
33
Fuhrmann
M.
,
Bittner
T.
,
Jung
C. K.
,
Burgold
S.
,
Page
R. M.
,
Mitteregger
G.
,
Haass
C.
,
LaFerla
F. M.
,
Kretzschmar
H.
,
Herms
J.
.
2010
.
Microglial Cx3cr1 knockout prevents neuron loss in a mouse model of Alzheimer’s disease.
Nat. Neurosci.
13
:
411
413
.
34
Kim
K. W.
,
Vallon-Eberhard
A.
,
Zigmond
E.
,
Farache
J.
,
Shezen
E.
,
Shakhar
G.
,
Ludwig
A.
,
Lira
S. A.
,
Jung
S.
.
2011
.
In vivo structure/function and expression analysis of the CX3C chemokine fractalkine.
Blood
118
:
e156
e167
.
35
Durkan
A. M.
,
Alexander
R. T.
,
Liu
G. Y.
,
Rui
M.
,
Femia
G.
,
Robinson
L. A.
.
2007
.
Expression and targeting of CX3CL1 (fractalkine) in renal tubular epithelial cells.
J. Am. Soc. Nephrol.
18
:
74
83
.
36
Serbina
N. V.
,
Pamer
E. G.
.
2006
.
Monocyte emigration from bone marrow during bacterial infection requires signals mediated by chemokine receptor CCR2.
Nat. Immunol.
7
:
311
317
.
37
Jia
T.
,
Serbina
N. V.
,
Brandl
K.
,
Zhong
M. X.
,
Leiner
I. M.
,
Charo
I. F.
,
Pamer
E. G.
.
2008
.
Additive roles for MCP-1 and MCP-3 in CCR2-mediated recruitment of inflammatory monocytes during Listeria monocytogenes infection.
J. Immunol.
180
:
6846
6853
.
38
Engel
D. R.
,
Maurer
J.
,
Tittel
A. P.
,
Weisheit
C.
,
Cavlar
T.
,
Schumak
B.
,
Limmer
A.
,
van Rooijen
N.
,
Trautwein
C.
,
Tacke
F.
,
Kurts
C.
.
2008
.
CCR2 mediates homeostatic and inflammatory release of Gr1high monocytes from the bone marrow, but is dispensable for bladder infiltration in bacterial urinary tract infection.
J. Immunol.
181
:
5579
5586
.
39
Landsman
L.
,
Jung
S.
.
2007
.
Lung macrophages serve as obligatory intermediate between blood monocytes and alveolar macrophages.
J. Immunol.
179
:
3488
3494
.
40
Lin
S. L.
,
Castaño
A. P.
,
Nowlin
B. T.
,
Lupher
M. L.
 Jr.
,
Duffield
J. S.
.
2009
.
Bone marrow Ly6Chigh monocytes are selectively recruited to injured kidney and differentiate into functionally distinct populations.
J. Immunol.
183
:
6733
6743
.
41
Krause
T. A.
,
Alex
A. F.
,
Engel
D. R.
,
Kurts
C.
,
Eter
N.
.
2014
.
VEGF-production by CCR2-dependent macrophages contributes to laser-induced choroidal neovascularization.
PLoS One
9
:
e94313
.
42
Schmidt
M.
,
Sun
G.
,
Stacey
M. A.
,
Mori
L.
,
Mattoli
S.
.
2003
.
Identification of circulating fibrocytes as precursors of bronchial myofibroblasts in asthma.
J. Immunol.
171
:
380
389
.
43
Pérez de Lema
G.
,
Maier
H.
,
Franz
T. J.
,
Escribese
M.
,
Chilla
S.
,
Segerer
S.
,
Camarasa
N.
,
Schmid
H.
,
Banas
B.
,
Kalaydjiev
S.
, et al
.
2005
.
Chemokine receptor Ccr2 deficiency reduces renal disease and prolongs survival in MRL/lpr lupus-prone mice.
J. Am. Soc. Nephrol.
16
:
3592
3601
.
44
Li
L.
,
Huang
L.
,
Sung
S. S.
,
Vergis
A. L.
,
Rosin
D. L.
,
Rose
C. E.
 Jr.
,
Lobo
P. I.
,
Okusa
M. D.
.
2008
.
The chemokine receptors CCR2 and CX3CR1 mediate monocyte/macrophage trafficking in kidney ischemia-reperfusion injury.
Kidney Int.
74
:
1526
1537
.
45
Tacke
F.
,
Alvarez
D.
,
Kaplan
T. J.
,
Jakubzick
C.
,
Spanbroek
R.
,
Llodra
J.
,
Garin
A.
,
Liu
J.
,
Mack
M.
,
van Rooijen
N.
, et al
.
2007
.
Monocyte subsets differentially employ CCR2, CCR5, and CX3CR1 to accumulate within atherosclerotic plaques.
J. Clin. Invest.
117
:
185
194
.
46
Auffray
C.
,
Fogg
D.
,
Garfa
M.
,
Elain
G.
,
Join-Lambert
O.
,
Kayal
S.
,
Sarnacki
S.
,
Cumano
A.
,
Lauvau
G.
,
Geissmann
F.
.
2007
.
Monitoring of blood vessels and tissues by a population of monocytes with patrolling behavior.
Science
317
:
666
670
.
47
Combadière
C.
,
Feumi
C.
,
Raoul
W.
,
Keller
N.
,
Rodéro
M.
,
Pézard
A.
,
Lavalette
S.
,
Houssier
M.
,
Jonet
L.
,
Picard
E.
, et al
.
2007
.
CX3CR1-dependent subretinal microglia cell accumulation is associated with cardinal features of age-related macular degeneration.
J. Clin. Invest.
117
:
2920
2928
.
48
Clover
A. J.
,
Kumar
A. H.
,
Caplice
N. M.
.
2011
.
Deficiency of CX3CR1 delays burn wound healing and is associated with reduced myeloid cell recruitment and decreased sub-dermal angiogenesis.
Burns
37
:
1386
1393
.
49
Furuichi
K.
,
Gao
J. L.
,
Murphy
P. M.
.
2006
.
Chemokine receptor CX3CR1 regulates renal interstitial fibrosis after ischemia-reperfusion injury.
Am. J. Pathol.
169
:
372
387
.
50
Karlmark
K. R.
,
Zimmermann
H. W.
,
Roderburg
C.
,
Gassler
N.
,
Wasmuth
H. E.
,
Luedde
T.
,
Trautwein
C.
,
Tacke
F.
.
2010
.
The fractalkine receptor CX3CR1 protects against liver fibrosis by controlling differentiation and survival of infiltrating hepatic monocytes.
Hepatology
52
:
1769
1782
.
51
Aoyama
T.
,
Inokuchi
S.
,
Brenner
D. A.
,
Seki
E.
.
2010
.
CX3CL1-CX3CR1 interaction prevents carbon tetrachloride-induced liver inflammation and fibrosis in mice.
Hepatology
52
:
1390
1400
.
52
Arai
M.
,
Ikawa
Y.
,
Chujo
S.
,
Hamaguchi
Y.
,
Ishida
W.
,
Shirasaki
F.
,
Hasegawa
M.
,
Mukaida
N.
,
Fujimoto
M.
,
Takehara
K.
.
2013
.
Chemokine receptors CCR2 and CX3CR1 regulate skin fibrosis in the mouse model of cytokine-induced systemic sclerosis.
J. Dermatol. Sci.
69
:
250
258
.
53
Zhang
J.
,
Patel
J. M.
.
2010
.
Role of the CX3CL1-CX3CR1 axis in chronic inflammatory lung diseases.
Int. J. Clin. Exp. Med.
3
:
233
244
.
54
Shimizu
K.
,
Furuichi
K.
,
Sakai
N.
,
Kitagawa
K.
,
Matsushima
K.
,
Mukaida
N.
,
Kaneko
S.
,
Wada
T.
.
2011
.
Fractalkine and its receptor, CX3CR1, promote hypertensive interstitial fibrosis in the kidney.
Hypertens. Res.
34
:
747
752
.
55
Ishida
Y.
,
Gao
J. L.
,
Murphy
P. M.
.
2008
.
Chemokine receptor CX3CR1 mediates skin wound healing by promoting macrophage and fibroblast accumulation and function.
J. Immunol.
180
:
569
579
.
56
Soos
T. J.
,
Sims
T. N.
,
Barisoni
L.
,
Lin
K.
,
Littman
D. R.
,
Dustin
M. L.
,
Nelson
P. J.
.
2006
.
CX3CR1+ interstitial dendritic cells form a contiguous network throughout the entire kidney.
Kidney Int.
70
:
591
596
.
57
Feng
L.
,
Chen
S.
,
Garcia
G. E.
,
Xia
Y.
,
Siani
M. A.
,
Botti
P.
,
Wilson
C. B.
,
Harrison
J. K.
,
Bacon
K. B.
.
1999
.
Prevention of crescentic glomerulonephritis by immunoneutralization of the fractalkine receptor CX3CR1 rapid communication.
Kidney Int.
56
:
612
620
.
58
Lionakis
M. S.
,
Swamydas
M.
,
Fischer
B. G.
,
Plantinga
T. S.
,
Johnson
M. D.
,
Jaeger
M.
,
Green
N. M.
,
Masedunskas
A.
,
Weigert
R.
,
Mikelis
C.
, et al
.
2013
.
CX3CR1-dependent renal macrophage survival promotes Candida control and host survival.
J. Clin. Invest.
123
:
5035
5051
.
59
Koziolek
M. J.
,
Schmid
H.
,
Cohen
C. D.
,
Blaschke
S.
,
Hemmerlein
B.
,
Zapf
A.
,
Müller
G. A.
,
Strutz
F.
.
2007
.
Potential role of fractalkine receptor expression in human renal fibrogenesis.
Kidney Int.
72
:
599
607
.
60
Lindquist
R. L.
,
Shakhar
G.
,
Dudziak
D.
,
Wardemann
H.
,
Eisenreich
T.
,
Dustin
M. L.
,
Nussenzweig
M. C.
.
2004
.
Visualizing dendritic cell networks in vivo.
Nat. Immunol.
5
:
1243
1250
.
61
Jung
S.
,
Aliberti
J.
,
Graemmel
P.
,
Sunshine
M. J.
,
Kreutzberg
G. W.
,
Sher
A.
,
Littman
D. R.
.
2000
.
Analysis of fractalkine receptor CX(3)CR1 function by targeted deletion and green fluorescent protein reporter gene insertion.
Mol. Cell. Biol.
20
:
4106
4114
.
62
Kuligowski
M. P.
,
Kitching
A. R.
,
Hickey
M. J.
.
2006
.
Leukocyte recruitment to the inflamed glomerulus: a critical role for platelet-derived P-selectin in the absence of rolling.
J. Immunol.
176
:
6991
6999
.
63
Devi
S.
,
Li
A.
,
Westhorpe
C. L.
,
Lo
C. Y.
,
Abeynaike
L. D.
,
Snelgrove
S. L.
,
Hall
P.
,
Ooi
J. D.
,
Sobey
C. G.
,
Kitching
A. R.
,
Hickey
M. J.
.
2013
.
Multiphoton imaging reveals a new leukocyte recruitment paradigm in the glomerulus.
Nat. Med.
19
:
107
112
.
64
Boor
P.
,
Konieczny
A.
,
Villa
L.
,
Schult
A. L.
,
Bücher
E.
,
Rong
S.
,
Kunter
U.
,
van Roeyen
C. R.
,
Polakowski
T.
,
Hawlisch
H.
, et al
.
2007
.
Complement C5 mediates experimental tubulointerstitial fibrosis.
J. Am. Soc. Nephrol.
18
:
1508
1515
.
65
Inoue
A.
,
Hasegawa
H.
,
Kohno
M.
,
Ito
M. R.
,
Terada
M.
,
Imai
T.
,
Yoshie
O.
,
Nose
M.
,
Fujita
S.
.
2005
.
Antagonist of fractalkine (CX3CL1) delays the initiation and ameliorates the progression of lupus nephritis in MRL/lpr mice.
Arthritis Rheum.
52
:
1522
1533
.
66
Machida
Y.
,
Kitamoto
K.
,
Izumi
Y.
,
Shiota
M.
,
Uchida
J.
,
Kira
Y.
,
Nakatani
T.
,
Miura
K.
.
2010
.
Renal fibrosis in murine obstructive nephropathy is attenuated by depletion of monocyte lineage, not dendritic cells.
J. Pharmacol. Sci.
114
:
464
473
.
67
Hammad
H.
,
Lambrecht
B. N.
.
2008
.
Dendritic cells and epithelial cells: linking innate and adaptive immunity in asthma.
Nat. Rev. Immunol.
8
:
193
204
.
68
Geissmann
F.
,
Jung
S.
,
Littman
D. R.
.
2003
.
Blood monocytes consist of two principal subsets with distinct migratory properties.
Immunity
19
:
71
82
.
69
Yanagita
M.
2012
.
Inhibitors/antagonists of TGF-β system in kidney fibrosis.
Nephrol. Dial. Transplant.
27
:
3686
3691
.
70
Martinez-Pomares
L.
,
Reid
D. M.
,
Brown
G. D.
,
Taylor
P. R.
,
Stillion
R. J.
,
Linehan
S. A.
,
Zamze
S.
,
Gordon
S.
,
Wong
S. Y.
.
2003
.
Analysis of mannose receptor regulation by IL-4, IL-10, and proteolytic processing using novel monoclonal antibodies.
J. Leukoc. Biol.
73
:
604
613
.
71
Xue
J.
,
Schmidt
S. V.
,
Sander
J.
,
Draffehn
A.
,
Krebs
W.
,
Quester
I.
,
De Nardo
D.
,
Gohel
T. D.
,
Emde
M.
,
Schmidleithner
L.
, et al
.
2014
.
Transcriptome-based network analysis reveals a spectrum model of human macrophage activation.
Immunity
40
:
274
288
.
72
Jenkins
S. J.
,
Ruckerl
D.
,
Cook
P. C.
,
Jones
L. H.
,
Finkelman
F. D.
,
van Rooijen
N.
,
MacDonald
A. S.
,
Allen
J. E.
.
2011
.
Local macrophage proliferation, rather than recruitment from the blood, is a signature of TH2 inflammation.
Science
332
:
1284
1288
.
73
Jenkins
S. J.
,
Ruckerl
D.
,
Thomas
G. D.
,
Hewitson
J. P.
,
Duncan
S.
,
Brombacher
F.
,
Maizels
R. M.
,
Hume
D. A.
,
Allen
J. E.
.
2013
.
IL-4 directly signals tissue-resident macrophages to proliferate beyond homeostatic levels controlled by CSF-1.
J. Exp. Med.
210
:
2477
2491
.
74
Landsman
L.
,
Bar-On
L.
,
Zernecke
A.
,
Kim
K. W.
,
Krauthgamer
R.
,
Shagdarsuren
E.
,
Lira
S. A.
,
Weissman
I. L.
,
Weber
C.
,
Jung
S.
.
2009
.
CX3CR1 is required for monocyte homeostasis and atherogenesis by promoting cell survival.
Blood
113
:
963
972
.
75
Mionnet
C.
,
Buatois
V.
,
Kanda
A.
,
Milcent
V.
,
Fleury
S.
,
Lair
D.
,
Langelot
M.
,
Lacoeuille
Y.
,
Hessel
E.
,
Coffman
R.
, et al
.
2010
.
CX3CR1 is required for airway inflammation by promoting T helper cell survival and maintenance in inflamed lung.
Nat. Med.
16
:
1305
1312
.
76
Lesnik
P.
,
Haskell
C. A.
,
Charo
I. F.
.
2003
.
Decreased atherosclerosis in CX3CR1‑/‑ mice reveals a role for fractalkine in atherogenesis.
J. Clin. Invest.
111
:
333
340
.
77
Dong
X.
,
Swaminathan
S.
,
Bachman
L. A.
,
Croatt
A. J.
,
Nath
K. A.
,
Griffin
M. D.
.
2007
.
Resident dendritic cells are the predominant TNF-secreting cell in early renal ischemia-reperfusion injury.
Kidney Int.
71
:
619
628
.
78
Lech
M.
,
Avila-Ferrufino
A.
,
Allam
R.
,
Segerer
S.
,
Khandoga
A.
,
Krombach
F.
,
Garlanda
C.
,
Mantovani
A.
,
Anders
H. J.
.
2009
.
Resident dendritic cells prevent postischemic acute renal failure by help of single Ig IL-1 receptor-related protein.
J. Immunol.
183
:
4109
4118
.
79
Tittel
A. P.
,
Heuser
C.
,
Ohliger
C.
,
Llanto
C.
,
Yona
S.
,
Hämmerling
G. J.
,
Engel
D. R.
,
Garbi
N.
,
Kurts
C.
.
2012
.
Functionally relevant neutrophilia in CD11c diphtheria toxin receptor transgenic mice.
Nat. Methods
9
:
385
390
.
80
Chorro
L.
,
Sarde
A.
,
Li
M.
,
Woollard
K. J.
,
Chambon
P.
,
Malissen
B.
,
Kissenpfennig
A.
,
Barbaroux
J. B.
,
Groves
R.
,
Geissmann
F.
.
2009
.
Langerhans cell (LC) proliferation mediates neonatal development, homeostasis, and inflammation-associated expansion of the epidermal LC network.
J. Exp. Med.
206
:
3089
3100
.

The authors have no financial conflicts of interest.