Introduction
Chemokines comprise a family of small chemoattractant proteins that play important roles in diverse host processes including chemotaxis, immune cell development, and leukocyte activation (Zlotnik and Yoshie, 2000; Zlotnik et al., 2011; Charo and Ransohoff, 2006). The chemokine superfamily includes 48 human ligands and 19 receptors, classified into subfamilies (CC, CXC, C, and CX3C) depending on the location of the cysteines in their sequence (Nomiyama et al., 2013; Hughes and Nibbs, 2018). Four chemokines predominate in mucosal tissues: CCL25, CCL28, CXCL14, and CXCL17 (Hernández-Ruiz and Zlotnik, 2017).
CCL28, also known as Mucosae-associated Epithelial Chemokine, belongs to the CC (or β-chemokine) subclass, and is constitutively produced in mucosal tissues including the digestive system, respiratory tract, and female reproductive system (Mohan et al., 2017). Although best studied for its homeostatic functions, CCL28 can also be induced under inflammatory conditions and is thus considered a dual function chemokine (Mohan et al., 2017).
CCL28 signals via two receptors: CCR3 and CCR10 (Pan et al., 2000). During homeostasis in mice, CCL28 provides a chemotactic gradient for CCR10+ B and T cells and guides the migration of CCR10+ IgA plasmablasts to the mammary gland and other tissues (Mohan et al., 2017; Burkhardt et al., 2019; Matsuo et al., 2018). In a disease context, CCL28 has been best studied in allergic airway inflammation. High CCL28 levels are present in airway biopsies from asthma patients (O’Gorman et al., 2005), and CCR3+ and CCR10+ cells are recruited to the airways in a CCL28-dependent fashion in murine asthma models (John et al., 2005; English et al., 2006).
In the human gut, CCL28 is upregulated during inflammation of the gastric mucosa in
Collectively, a variety of studies have postulated that CCL28 is an important chemokine in inflammatory diseases, ranging from asthma to ulcerative colitis, and during the immune response to infection. Yet, CCL28 function remains understudied, largely because
By comparing infection in
Results
CCL28-mediated responses limit
We investigated CCL28 activity during gastrointestinal infection with STm by using the well-established streptomycin-treated C57BL/6 mouse model of colitis (Barthel et al., 2003; Walker et al., 2023). At day 4 post-infection (4 dpi) with STm, we observed a ~fourfold increase of CCL28 by enzyme-linked immunosorbent assay (ELISA) analysis of feces from wild-type mice relative to uninfected controls (Figure 1A). In a prior preliminary study, we found that
Figure 1.
CCL28 confers protection during
(A) For the colitis model, wild-type (WT) mice were gavaged with streptomycin 24 hr prior to oral infection with approximately 1 × 109 CFU
Figure 1—figure supplement 1.
(A) STm CFU in the fecal content collected 1 and 2 dpi, and in the cecal content 2 dpi from wild-type (WT, filled circles) and
Figure 1—figure supplement 2.
CCL28 does not confer protection in a
(A, B) For the bacteremia model, mice were infected by intraperitoneal injection with
Figure 1—figure supplement 3.
Flow cytometry gating strategy for the identification and classification of major immune cell populations in the tissues of STm-infected mice.
An equivalent strategy was also used to immunophenotype cells from uninfected and
Figure 1—figure supplement 4.
Wild-type (WT) and
Flow cytometry quantification of live, CD45+ CD11b− immune cells recovered from WT and
Figure 1—figure supplement 5.
Profiling granulocyte and APC-like cell abundance in wild-type (WT) and
Flow cytometry quantification of live, CD45+ CD11b+ immune cells recovered from WT and
Figure 1—figure supplement 6.
Neutrophil-associated antimicrobial protein levels during intestinal STm infection of wild-type (WT) and
The levels of myeloperoxidase (MPO; A) neutrophil elastase (B), and S100A9 (a component of the antimicrobial calcium-binding protein calprotectin; C), were measured by ELISA from the fecal and cecal supernatant of STm-infected WT and
CCL28 promotes neutrophil accumulation to the gut during
CCL28 has direct antimicrobial activity against some bacteria (e.g.,
During homeostasis, CCL28 exhibits chemotactic activity in the gut mucosa toward CD4+ and CD8+ T cells and IgA-producing B cells (Mohan et al., 2017; Burkhardt et al., 2019; Matsuo et al., 2018). However, immune cell profiling in the intestines (using the flow cytometry gating strategy presented in Figure 1—figure supplement 3) revealed similar B cell and CD4+ and CD8+ T cell numbers in both wild-type and
We detected slightly lower levels of the NET-associated peptides myeloperoxidase (MPO), neutrophil elastase, and S100A9 (a subunit of calprotectin, a metal-sequestering protein associated with neutrophils) in the cecal content supernatant of STm-infected
Gut proinflammatory gene expression and tissue pathology are reduced in
Neutrophils can mediate inflammation by producing proinflammatory molecules or engaging in crosstalk with other cells (Sabroe et al., 2005). We evaluated the expression of genes encoding proinflammatory cytokines in the cecum of
CCL28 is expressed in several mucosal tissues beyond the gut, including the lung (Mohan et al., 2017). To investigate whether CCL28 promotes neutrophil accumulation and host protection in the lung, we employed a murine Ab pneumonia model (Dillon et al., 2019; Lin et al., 2015). Ab is an emerging, frequently multidrug-resistant Gram-negative pathogen causing potentially lethal nosocomial pneumonia (Ayoub Moubareck and Hammoudi Halat, 2020). Following intratracheal Ab infection, we observed a striking phenotype: 75% of wild-type mice died within 48 hr, whereas 88% of
Figure 2.
Absence of CCL28 confers protection in a lethal
(A) Wild-type (WT) mice (solid black line) and
Figure 2—figure supplement 1.
Immunophenotyping of CD11b+ immune cells recovered from wild-type (WT) and
Data indicate the relative abundance of neutrophils (A, CD11b+ Ly6G+), eosinophils (B, CD11b+ Ly6G− SiglecF+ side scatterhigh), macrophage-like F4/80+ CD11c− cells (C), CD11b+ Ly6G− SiglecF− F4/80+ CD11c−, and conventional dendritic cell-like CD11c+ F4/80 cells (D), CD11b+ Ly6G− SiglecF− CD11c+ F4/80− as proportions of total live CD45+ cells in the bronchoalveolar lavage (BAL), lungs, blood, and bone marrow, from uninfected (naive) and 1 day post-inoculation with
Figure 2—figure supplement 2.
Immunophenotyping of lymphocytes recovered from wild-type (WT) and
Data indicate the relative abundance of B cells (A, CD11b− CD3− CD19+), CD8+ T cells (B, CD11b− CD19− CD3+ CD4− CD8+), and CD4+ T cells (C), CD11b− CD19− CD3+ CD4+ CD8−, as proportions of total live CD45+ cells in the bronchoalveolar lavage (BAL), lungs, blood, and bone marrow, from uninfected (naive) and 1 dpi with
Figure 2—figure supplement 3.
Neutrophil-associated antimicrobial protein levels during lung Ab infection of wild-type (WT) and
The levels of myeloperoxidase (MPO; A) neutrophil elastase (B), and S100A9 (C), were measured by ELISA from the supernatant of the bronchoalveolar lavage fluid (BAL) from uninfected WT and Ab-infected WT and
In vitro, high concentrations (1 μM) of CCL28 exhibited direct antimicrobial activity against 5 × 105 CFU of Ab, but not when higher CFU (5 × 108/ml) were used as inoculum in the assay (Figure 1—figure supplement 2C). Given that high Ab CFU were recovered in the lung of wild-type mice (Figure 2B, C), CCL28 does not appear to limit growth of this pathogen in vivo even though it exhibits modest antimicrobial activity in vitro. We thus investigated if alterations in neutrophil accumulation in the lung between wild-type and
CCL28 promotes neutrophil accumulation to the lung during
Prior studies demonstrated neutrophil recruitment to the lungs of Ab-infected mice beginning at 4 hr post-infection and peaking at 1 dpi (van Faassen et al., 2007; Tsuchiya et al., 2012). CCL28 contributed to neutrophil recruitment during STm gut infection, so we analyzed neutrophil recruitment to the lung mucosa 1 day after Ab infection in wild-type and
Gut and BAL neutrophils express receptors CCR3 and CCR10 during infection
CCL28 attracts leukocytes expressing at least one of its receptors, CCR3 or CCR10. CCR10 is found on T cells, B cells, and IgA-secreting plasma cells, whereas eosinophils express CCR3 (Mohan et al., 2017). Although early studies concluded that CCR3 was absent in neutrophils (Höchstetter et al., 2000), later research detected this receptor on neutrophils isolated from patients with chronic inflammation (Hartl et al., 2008). Based on these findings and our observations of CCL28-dependent neutrophil accumulation in the gut during STm colitis and in the lung during Ab infection (Figures 1 and 2), we performed flow cytometry on single-cell suspensions from infected mouse tissues to evaluate surface expression of CCR3 and CCR10. In STm-infected mice, we analyzed the gut, blood, and bone marrow (Figure 3A, B). Both receptors were present on a small subset of bone marrow neutrophils (~4% CCR3, ~0.2% CCR10) and blood neutrophils (~5% CCR3, ~1% CCR10) during infection. However, neutrophils expressing these receptors, particularly CCR3, were enriched in the inflamed gut, with ~20% expressing CCR3 and ~2% expressing CCR10 (Figure 3A, B). Simultaneously staining for both CCR3 and CCR10 showed that ~1% of gut neutrophils from infected wild-type mice expressed both receptors (Figure 3—figure supplement 1A), and infected
Figure 3.
Surface expression of the CCL28 receptors CCR3 and CCR10 on neutrophils from infected tissue, and upon stimulation with proinflammatory stimuli and phagocytosis.
Surface expression of (A, C) CCR3 or (B, D) CCR10 on murine neutrophils obtained from (A, B) the gut, blood, and bone marrow (BM) 3 dpi with STm, or (C, D) the bronchoalveolar lavage (BAL), blood, and bone marrow 1 dpi with Ab, analyzed by flow cytometry. Left panels show representative histograms of (A, C) CCR3 or (B, D) CCR10 expression on the surface of neutrophils (gated on live, CD45+ CD11b+ Ly6G+ cells) from (A, B) the gut (blue), blood (red), and bone marrow (BM; black) or (C, D) BAL (blue), blood (red), and bone marrow (BM; black). Right panels show the percentage of (A, C) CCR3+ or (B, D) CCR10+ neutrophils obtained from (A, B) gut, blood, and BM or (C, D) BAL, blood, and BM. Data are from six independent experiments. (E–H) Uninfected bone marrow neutrophils were unstimulated or treated with the indicated stimuli for 4 hr. Surface expression of (E, G) CCR3 and (F, H) CCR10 on neutrophils was determined by flow cytometry. Left panels show representative histograms of (E, G) CCR3 or (F, H) CCR10 surface expression after stimulation with: (E, F) cytokines IFNγ + TNFɑ + GM-CSF (blue); fMLP (magenta); phorbol 12-myristate 13-acetate (PMA) (purple); lipopolysaccharide (LPS) (red); (G, H) cytokines IFNγ + TNFɑ + Granulocyte-macrophage colony stimulating factor (GM-CSF, blue); beads alone (magenta); cytokines plus beads (red). Right panels show the percentage of (E, G) CCR3+ or (F, H) CCR10+ neutrophils following stimulation with the indicated stimuli. US = unstimulated. Data shown are pooled from two independent experiments. (I, J) Bone marrow cells enriched for neutrophils were infected with opsonized STm at a multiplicity of infection (MOI) = 10 for 1 hr with (violet) or without (red) pretreatment with cytochalasin D for 30 min before infection. Surface expression of (I) CCR3 or (J) CCR10 was determined by flow cytometry. Data are from two independent experiments. Left panels show representative histograms of surface receptor staining on neutrophils, and right panels show the percentages. (A–J, right panels) Bars represent the mean ± standard deviation (SD). (A–D) Data were analyzed by one-way analysis of variance (ANOVA) for paired samples (non-parametric Friedman test), assuming non-normal distribution and non-equal SD given the differences in the variance among the groups, followed by Dunn’s multiple comparisons test. (E–J) Data were analyzed by one-way ANOVA for paired samples, applying the Greenhouse–Geisser correction given the differences in variance among the groups; Bonferroni’s multiple comparison test was performed to compare between relevant stimulation conditions. Significant changes are indicated by *p ≤ 0.05, **p ≤ 0.01, ***p ≤ 0.001, ****p ≤ 0.0001; ns, not significant.
Figure 3—figure supplement 1.
Expression of CCR3 and CCR10 in neutrophils isolated from the gut and lung mucosa in infected wild-type (WT) and
(A) Surface expression of CCR3 and CCR10 on neutrophils obtained from the gut of WT mice (
Neutrophils isolated from the BAL of Ab-infected wild-type mice also expressed CCR3 and CCR10 surface expression, with ~15% of neutrophils expressing CCR3 (Figure 3C) and ~2% expressing CCR10 (Figure 3D). Simultaneously staining for both CCR3 and CCR10 revealed that ~0.5% of BAL neutrophils from infected wild-type mice expressed both receptors (Figure 3—figure supplement 1C), and infected
Proinflammatory stimuli and phagocytosis induce expression of CCR3 and CCR10 on neutrophils
We investigated mechanisms underpinning the upregulation of CCR3 and CCR10 in neutrophils. A prior study indicated that a cocktail of proinflammatory cytokines (GM-CSF, IFNγ, TNFɑ) boosts CCR3 expression in human peripheral blood neutrophils from healthy donors (Hartl et al., 2008), and expression of these cytokines is highly induced during STm colitis (Figure 1F) and Ab pneumonia (Figure 2L). We stimulated bone marrow neutrophils from wild-type mice (which express low levels of CCR3 and CCR10) with these cytokines, and independently with other pro-inflammatory compounds including lipopolysaccharide (LPS), the protein kinase C activator phorbol 12-myristate 13-acetate (PMA), or the
Phagocytosis of microbes and necrotic debris are critical neutrophil functions at tissue foci of infection and inflammation (Uribe-Querol and Rosales, 2020) and are associated with changes in neutrophil gene expression (Kobayashi et al., 2002). We tested whether phagocytosis induced CCR3 and CCR10 expression by incubating bone marrow neutrophils with latex beads, with or without the cytokine cocktail. Phagocytosis of latex beads alone resulted in a small but significant induction of neutrophil CCR3 expression (~8% of neutrophils); however, latex beads augmented with the cytokine cocktail markedly induced CCR3 expression (~25% of neutrophils vs. ~15% with cocktail alone; Figure 3G). This synergistic effect of phagocytosis was not notable for CCR10 (Figure 3H).
To further probe the role of phagocytosis in CCR3 expression, we incubated bone marrow neutrophils with live STm for 1 hr. STm rapidly induced CCR3 expression on the neutrophil surface (~25% of cells; Figure 3I), whereas CCR10 was only minimally induced (Figure 3J). Cytochalasin D, a potent inhibitor of the actin polymerization required for phagocytic uptake, largely blocked CCR3 receptor induction (Figure 3I); however, CCR10 induction was not blocked (Figure 3J), suggesting that a mechanism other than phagocytic uptake likely drives the minor increase in CCR10 expression by neutrophils. Incubation of bone marrow neutrophils with CCL28 (both alone and in the context of STm co-incubation) had negligible effects on CCR3 and CCR10 levels (data not shown). Thus, proinflammatory stimuli and phagocytosis enhance CCR3 and, to a lesser extent, CCR10 expression on the neutrophil surface.
CCR3 is stored intracellularly in neutrophils
Neutrophil intracellular compartments and granules harbor enzymes, cytokines, and receptors necessary for rapid responses to pathogens. For example, activation of human neutrophils induces rapid translocation of complement receptor type 1 (CR1) from an intracellular compartment to the cell surface, increasing its surface expression up to 10-fold (Berger et al., 1991). Given the rapid (within 1 hr) increase of neutrophil CCR3 surface expression upon STm infection, we hypothesized that CCR3, akin to CR1, may be stored intracellularly in neutrophils, consistent with reports of intracellular CCR3 in eosinophils (Spencer et al., 2006).
Uninfected bone marrow neutrophils maintained relatively low surface levels of CCR3 (Figure 4A), but when permeabilized for intracellular staining, almost all (~99%) were CCR3+, indicating intracellular storage (Figure 4B). Upon STm infection in vitro, bone marrow neutrophils increased CCR3 surface expression as quickly as 5 min post-infection, reaching a maximum of ~30% CCR3+ neutrophils at 2 hpi (Figure 4A). These results suggest mobilization of pre-formed receptor from an intracellular compartment (Figure 4B). Intracellular stores of CCR10 were also detected in some bone marrow neutrophils under homeostatic conditions, with a small but significant increase during STm infection (Figure 4—figure supplement 1B). However, CCR10 was expressed on the surface of only ~0.3% uninfected bone marrow neutrophils, increasing to ~0.6% during STm infection (Figure 4—figure supplement 1A). In vitro, Ab infection induced less CCR3 surface expression on neutrophils relative to STm (~7–10%) and took longer to observe the increased CCR3+ staining (Figure 4C), whereas CCR10 did not significantly increase (Figure 4—figure supplement 1C). Most bone marrow neutrophils also expressed intracellular CCR3 (Figure 4D) and CCR10 (Figure 4—figure supplement 1D) during Ab infection. Similar findings were observed in neutrophils isolated from bone marrow, blood, and gut tissue of mice orally infected with STm, and from bone marrow, blood, and BAL fluid of mice infected with Ab, with both intracellular and surface CCR3 observed (Figure 4E, F). CCR3 surface expression levels were higher on neutrophils isolated from the gut relative to other sites (Figure 4E), though levels in the BAL fluid were similar to Ab-infected blood and bone marrow neutrophils (Figure 4F). Neutrophils expressing surface CCR10 were low in all tissues, though slightly higher in the STm-infected gut than in blood and bone marrow, with intracellular stores of CCR10 also observed (Figure 4—figure supplement 1E, F). We conclude that CCR3 is stored intracellularly in neutrophils and rapidly mobilized to the cell surface upon infection, phagocytosis, and/or cytokine stimulation.
Figure 4.
Neutrophil CCR3 is stored in intracellular compartments and rapidly mobilizes to the cell surface during infection.
Neutrophils enriched from wild-type mouse bone marrow were infected at multiplicity of infection (MOI) = 10 for 5 min to 4 hr with (A, B) opsonized
Figure 4—figure supplement 1.
Expression kinetics of neutrophil CCR10.
Neutrophils enriched from wild-type mouse bone marrow were infected at multiplicity of infection (MOI) = for 5 min to 4 hr with (A, B) opsonized
CCL28 enhances neutrophil antimicrobial activity, ROS production, and NET formation via CCR3 stimulation
Chemokines are essential for neutrophil migration to infection sites and may regulate additional neutrophil bactericidal effector functions, including the production of ROS and formation of NETs (Capucetti et al., 2020). We tested if CCL28 has chemotactic and/or immunostimulatory activity toward bone marrow neutrophils in vitro after boosting their CCR3 surface expression with the cytokine cocktail (GM-CSF + IFNγ + TNFɑ) as shown in Figure 3. We incubated the neutrophils with CCL28, the well-known neutrophil chemoattractant CXCL1, or with CCL11/eotaxin, a chemokine that binds CCR3 and is induced in the asthmatic lung to promote eosinophil recruitment (Conroy and Williams, 2001; Garcia-Zepeda et al., 1996; Kitaura et al., 1996). We found that CCL28 promoted neutrophil chemotaxis, though not as potently as CXCL1, while CCL11 had no significant effect (Figure 5A).
Figure 5.
CCL28 enhances neutrophil antimicrobial activity.
(A) Murine bone marrow neutrophils were stimulated with IFNγ + TNFɑ + GM-CSF for 4 hr before adding 1 × 106 cells to the upper compartment of a transwell chamber for chemotaxis assays. Each of the chemokines (CCL28, CCL11, or CXCL1), or no chemokine (NC), was placed in separate lower compartments. The transwell plate was incubated for 2 hr at 37°C. Cells that migrated to the lower compartment were enumerated by flow cytometry. Neutrophil chemotaxis index was calculated by taking the number of cells that migrated in response to a chemokine and dividing it by the number of cells that migrated in the absence of a chemokine. Data are from four independent experiments. (B, C) Infection of bone marrow neutrophils. (B) Opsonized STm (1 × 107 CFU) or (C) opsonized Ab (1 × 107 CFU) were cultured alone, or added to bone marrow neutrophils (1 × 106 cells) stimulated with CCL28, CCL11, or no chemokine, for 2.5 hr (STm) or 4.5 hr (Ab) at 37°C. Neutrophils were lysed with 1% Triton-X and surviving bacteria were enumerated by plating serial dilutions. Percentage of bacterial survival was calculated for each condition by taking the CFU from bacteria incubated with neutrophils and dividing it by the CFU from bacteria incubated without neutrophils, multiplied by 100. Data shown for each infection comprise three independent experiments. Bars represent the mean ± standard deviation (SD). (D) The effect of the CCR3 antagonist SB328437 on neutrophil-mediated STm killing was evaluated by performing the experiment as described in panel (B), with or without the antagonist. Data shown comprise three independent experiments. (E–G) Reactive oxygen species (ROS) production (2′,7′-dichlorodihydrofluorescein diacetate [H2DCFDA] conversion to fluorescent DCF) detected by flow cytometry in bone marrow neutrophils infected with STm as described in panel (B). In (F, G), cells were stimulated with CCL28 in the presence of an anti-CCR3 antibody, an anti-CCR10 antibody, or isotype controls. Left panels show representative histograms, and right panels show the percentage of ROS+ neutrophils in the indicated treatment groups. (H, I) Neutrophil extracellular trap (NET) formation detected by fluorescence microscopy using Helix dye in human neutrophils activated with platelets. Cells were unstimulated (no chemokine, NC), stimulated with CCL28 alone, or with CCL28 and the CCR3 agonist SB328737 and/or the CCR10 agonist BI-6901, as indicated. (H) Representative images of fluorescence microscopy with DAPI (blue) and Helix (green). (I) Quantification of NETs represented as percentage of cells forming NETs based on observed morphology. Connected circles represent NET abundance in cell populations from the same donor following different indicated treatments. (A–E) Bars represent the mean ± SD. (A–C) Data were analyzed by non-parametric analysis of variance (ANOVA) (Kruskal–Wallis’s test), assuming non-equal SD given the differences in the variance among the groups, followed by Dunn’s multiple comparisons test. (D, I) Data were analyzed by ratio paired
Figure 5—figure supplement 1.
Neutrophil extracellular trap (NET) formation (Helix+ MPO+ neutrophils) detected by flow cytometry in human neutrophils activated with platelets.
As indicated, cells were unstimulated (NC), stimulated with CCL28 alone, or with CCL28 and the CCR3 antagonist SB328437 and/or the CCR10 antagonist BI-6901 (as in Figure 5H, I). (A) Representative contour plots, and (B) percentage of Helix+ MPO+ neutrophils in the indicated treatment groups. Connected circles represent NET abundance in cell populations from the same donor following different indicated treatments. Ratio paired
To test whether CCL28 stimulation enhanced neutrophil effector function, we incubated STm with bone marrow neutrophils for 2.5 hr with or without CCL28 (50 nM) or CCL11 (50 nM), then quantified bacterial killing. Stimulation with CCL28 significantly increased neutrophil bactericidal activity against STm, with ~40% of the bacterial inoculum cleared, compared to ~10% clearance by unstimulated neutrophils (Figure 5B). Neutrophils stimulated with CCL11 displayed an intermediate phenotype (~25% bacterial killing). Neither chemokine exhibited direct antimicrobial activity against STm (Figure 1—figure supplement 2D). In contrast, ex vivo neutrophil killing of Ab was not significantly enhanced by CCL28 or CCL11 treatment (Figure 5C). Thus, although CCL28 modulates neutrophil accumulation in the lung during Ab infection (Figure 2D–J), it fails to reduce pathogen burden in the lung (Figure 2B) likely because CCL28 stimulation does not enhance neutrophil bactericidal activity against Ab.
Our data indicate that CCR3 is the primary CCL28 receptor expressed in neutrophils during STm infection (Figure 3I and 4). We tested whether the CCL28-mediated increase in neutrophil bactericidal activity could be reversed using SB328437, a CCR3 antagonist (White et al., 2000). SB328437 reversed the effects of both CCL28 and CCL11 on neutrophils, confirming receptor specificity (Figure 5D). An important mechanism of bacterial killing is the production of ROS (Fang, 2011), which is triggered by infection and enhanced by proinflammatory stimuli including cytokines and chemokines (Nguyen et al., 2017). We measured ROS production by incubating neutrophils with the cell-permeable probe 2′,7′-dichlorodihydrofluorescein diacetate (H2DCFDA), which forms the fluorescent byproduct 2′,7′-dichorofluorescein (DCF) when oxidized by ROS, and found that CCL28 stimulation enhanced neutrophil ROS production during STm infection (Figure 5E). The increased ROS production triggered by CCL28 was reversed when neutrophils were incubated with an anti-CCR3 blocking antibody (Figure 5F), but not with an anti-CCR10 blocking antibody (Figure 5G).
In addition to their direct antimicrobial activity, ROS trigger other neutrophil responses, including NET formation (Nguyen et al., 2017). NETs can be induced by various stimuli, including microbial products, inflammatory cytokines and chemokines, immune complexes, and activated platelets (Boeltz et al., 2019). To determine whether CCL28 enhances NET formation, we incubated human neutrophils with activated platelets with or without CCL28, then incubated the cells with the DNA-staining dyes DAPI and HELIX, and evaluated NET formation by fluorescence microscopy (Figure 5H). Incubation with activated platelets and CCL28 increased the percentage of NETs compared to neutrophils not stimulated with CCL28 (Figure 5H, I). Complementary experiment, analyzing DNA–MPO complexes confirmed an increased percentage of DNA–MPO complexes in response to platelet and CCL28 stimulation (Figure 5—figure supplement 1). The effect of CCL28 on platelet-activated NET formation was primarily mediated by CCR3, as the CCR3 antagonist SB328437 significantly reduced the percentage of observable NET+ neutrophils (Figure 5H1) and DNA–MPO complexes (Figure 5—figure supplement 1). In contrast, the CCR10 antagonist BI-6901 did not significantly reduce NET formation, and combined antagonism of CCR3 and CCR10 had an effect similar to CCR3 antagonism alone (Figure 5H1, Figure 5—figure supplement 1). Together, these results demonstrate that CCL28 enhances neutrophil ROS production and NET formation primarily in a CCR3-dependent manner.
Discussion
The mucosal immune response serves to maintain tissue homeostasis and to protect the host against invading pathogens. Here, we discovered that the chemokine CCL28 significantly contributes to neutrophil accumulation and activation in the mucosa during gastrointestinal infection with
Consistent with our initial observation that
Neutrophils are a hallmark of inflammatory diarrhea and are rapidly recruited to the gut following infection in the
Although an initial study concluded CCR3 was absent on neutrophils (Höchstetter et al., 2000), subsequent studies reported CCR3 expression on human neutrophils isolated from patients with chronic lung disease (Hartl et al., 2008) and on neutrophils isolated from the BAL fluid of mice infected with influenza (Rudd et al., 2019). Our study demonstrates that a substantial number of neutrophils isolated from infected mucosal sites express CCR3, and fewer express CCR10 on their surface, while resting neutrophils do not express these receptors on their surface (Figure 3). The rapid surface expression of CCR3 on neutrophils upon infection suggests that the receptor is stored intracellularly, similar to eosinophils (Spencer et al., 2006). Indeed, neutrophils isolated from bone marrow, blood, and infected mucosal tissue were all positive for CCR3 intracellular staining (Figure 4). In vitro, we could recapitulate the increase in surface receptor expression by incubating bone marrow neutrophils with proinflammatory stimuli (LPS, or the cytokines GM-CSF + IFNγ + TNFɑ) or following phagocytosis of bacterial pathogens (Figure 3). CCL28 stimulation of bone marrow neutrophils in vitro increased their antimicrobial activity and ROS production during
A reduction of neutrophil accumulation was also observed in the BAL and lung of
Even though CCL28 exhibited direct antimicrobial activity against
Overall, this study demonstrates that CCL28 plays an important role in the mucosal response to pathogens by promoting neutrophil accumulation at the site of infection. Neutrophils isolated from infected mucosa express the CCL28 receptors CCR3 and CCR10, and CCL28 enhances neutrophil activation, ROS production, and NET formation, primarily through CCR3. These findings have implications for other infectious and non-infectious diseases where neutrophil recruitment plays a major role, and may lead to the identification of CCL28-targeted therapies to modulate neutrophil function and mitigate collateral damage.
Materials and methods
Key resources table
Reagent type (species) or resource | Designation | Source or reference | Identifiers | Additional information |
---|---|---|---|---|
Strain, strain background ( | Lab stock; PMID:7868611 | Nalidixic acid-resistant derivative of strain ATCC 14028s | ||
Strain, strain background ( | Lab stock; from Michael McClelland PMID:19578432 | PhoQ coding sequence disrupted by a kanamycin cassette | ||
Strain, strain background ( | Lab Stock | ATCC Cat#700926 | ||
Strain, strain background ( | Walter Reed Medical Center; PMID:24865555 | |||
Genetic reagent ( | C57BL/6 | Deltagen; PMID:30855201 | Obtained from Albert Zlotnik (UC Irvine); Allelic exchange into Ccl28 | |
Genetic reagent ( | C57BL/6 | Cyagen Biosciences | Product Number: S-KO-17095; | Generated by CRISPR/Cas9-mediated deletion of exons 1–3 |
Biological sample ( | Primary human blood neutrophils | Human volunteers, UNAM | Freshly isolated from human volunteers | |
Biological sample ( | Primary bone marrow cells | C57BL/6 | Freshly isolated from wild-type mice of the | |
Antibody | Anti-mouse CD16/CD32 | BioLegend | Clone: 93; Cat#101302; | FC (1:50) |
Antibody | Anti-mouse CD45 | BioLegend | Clone: 30-F11; Cat#103126; | Sony SA3800 FC (1:800); |
Antibody | Anti-mouse/human CD11b | BioLegend | Clone: M1/70; Cat#101290; | FC (1:400) |
Antibody | Anti-mouse Ly6G | BioLegend | Clone: 1A8; Cat#127628; | FC (1:1600) |
Antibody | Anti-mouse CD170 (SiglecF) | BioLegend | Clone: S17007L; Cat#155530; | FC (1:400) |
Antibody | Anti-mouse CCR3 | R&D Biosystems | Clone: 83103; Cat#FAB729P; | FC (1:100) |
Antibody | Anti-mouse CCR10 | R&D Biosystems | Clone: 248918; Cat#FAB2815; | FC (1:100) |
Antibody | Anti-mouse CD11c | BioLegend | Clone: N418; Cat#117343; | FC (1:400) |
Antibody | Anti-mouse Ly6G | BioLegend | Clone: 1A8; Cat#127606; | FC (1:400) |
Antibody | Anti-mouse CD170 (SiglecF) | BioLegend | Clone: S17007L; Cat#155503; | FC (1:400) |
Antibody | Anti-mouse F4/80 | BioLegend | Clone: BM8; Cat#123146; | FC (1:400) |
Antibody | Anti-mouse CD8a | BioLegend | Clone: 53-6.7; Cat#100737; | FC (1:1600) |
Antibody | Anti-mouse CD3 | BioLegend | Clone: 17A2; Cat#100204; | FC (1:400) |
Antibody | Anti-mouse CD4 | BioLegend | Clone: RM4-5; Cat#100539; | FC (1:800) |
Antibody | Anti-mouse CD8a | BioLegend | Clone: 53-6.7; Cat#100708; | FC (1:1600) |
Antibody | Anti-mouse CD19 | BioLegend | Clone: 6D5; Cat#115528; | FC (1:400) |
Antibody | Anti-mouse/human CD11b | BioLegend | Clone: M1/70; Cat#101212; | FC (1:800) |
Antibody | Anti-mouse/human CD11b | BioLegend | Clone: M1/70; Cat#101245; | FC (1:400) |
Antibody | Anti-mouse F4/80 | BioLegend | Clone: BM8; Cat#123108; | FC (1:200) |
Antibody | Anti-mouse Ly6G | BioLegend | Clone: 1A8; Cat#127654; | FC (1:400) |
Antibody | Anti-mouse CD170 (SiglecF) | BioLegend | Clone: S17007L; Cat#155508; | FC (1:400) |
Antibody | Anti-mouse CD11c | BioLegend | Clone: N418; Cat#117317; | FC (1:400) |
Antibody | Anti-mouse CD19 | BioLegend | Clone: 6D5; Cat#115520; | FC (1:400) |
Antibody | Anti-mouse CCR3 | R&D Systems | Clone: 83103; Cat#MAB1551; | In vitro signaling blockade |
Antibody | Anti-mouse CCR10 | R&D Systems | Clone: 248918; Cat#MAB2815; | In vitro signaling blockade |
Antibody | Rat IgG2A Isotype Control Antibody | R&D Systems | Clone: 54447; Cat#MAB006; | In vitro signaling blockade |
Antibody | Anti-mouse Ly6G | BioLegend | Clone: 1A8; Cat#127601; | Lung neutrophil IF (1:100) |
Antibody | Goat Anti-rat IgG (H+L) | Invitrogen | Cat#A-21434; | Lung neutrophil IF: (1:400) |
Antibody | Human TruStain FcX | BioLegend | Cat#422302; | FC (1:100) |
Antibody | Anti-human CD45 | BioLegend | Clone: HI30; Cat#304028; | FC (1:300) |
Antibody | Anti-mouse/human CD11b | BioLegend | Clone: M1/70; Cat#101224; | FC (1:200) |
Antibody | Anti-human CD62L | BioLegend | Clone: DREG-56; Cat#304838; | FC (1:300) |
Antibody | Anti-human CCR3 | R&D Systems | Clone: 61828; Cat#FAB155P; | FC (1:100) |
Antibody | Anti-human CCR10 | R&D Systems | Clone: 314305; Cat#FAB3478A; | FC (1:100) |
Antibody | Anti-human myeloperoxidase | Novus Biologicals | Clone MPO421-8B2; Cat#NBP2-41406B | FC (1:50) |
Sequence-based reagent | Mouse | IDT | Forward: | |
Sequence-based reagent | Mouse | IDT | Forward: | |
Sequence-based reagent | Mouse | IDT | Forward: | |
Sequence-based reagent | Mouse | IDT | Forward: | |
Sequence-based reagent | Mouse | IDT | Forward: | |
Sequence-based reagent | Mouse | IDT | Forward: | |
Sequence-based reagent | Mouse | IDT | Forward: | |
Peptide, recombinant protein | Recombinant Mouse CCL28 (MEC) | BioLegend | Cat#584706 | In vitro killing: various concentrations (indicated in text) |
Peptide, recombinant protein | Recombinant Mouse CCL28 Protein | R&D Systems | Cat#533-VI | Chemotaxis: 50 nM; neutrophil stimulation: 50 nM |
Peptide, recombinant protein | Recombinant Mouse CCL11/Eotaxin Protein | R&D Systems | Cat#420-ME | Chemotaxis: 50 nM; neutrophil stimulation: 25 nM |
Peptide, recombinant protein | Recombinant Murine KC (CXCL1) | Peprotech | Cat#250–11 | Chemotaxis: 50 nM |
Peptide, recombinant protein | Recombinant human CCL28 | BioLegend | Cat#584602 | Neutrophil stimulation: 50 nM |
Peptide, recombinant protein | Recombinant Mouse TNF-α | BioLegend | Cat#575202 | Neutrophil stimulation: 100 ng/ml |
Peptide, recombinant protein | Recombinant Mouse IFN-γ | BioLegend | Cat#575304 | Neutrophil stimulation: 500 U/ml |
Peptide, recombinant protein | Recombinant Mouse GM-CSF | BioLegend | Cat#576302 | Neutrophil stimulation: 10 ng/ml |
Peptide, recombinant protein | LPS-B5 Ultrapure | Invivogen | Cat#tlrl-pb5lps | Mouse neutrophil stimulation: 100 ng/ml |
Commercial assay or kit | EasySep Mouse Neutrophil Enrichment Kit | STEMCELL Technologies | Cat#19762 | |
Commercial assay or kit | EasySep Direct Human Neutrophil Isolation Kit | STEMCELL Technologies | Cat#19666 | |
Commercial assay or kit | Mouse CCL28 ELISA Max Deluxe | BioLegend | Cat# 441304 | |
Commercial assay or kit | Mouse Myeloperoxidase DuoSet ELISA Kit | R&D Systems | Cat#DY3667 | |
Commercial assay or kit | Mouse Neutrophil Elastase/ELA2 DuoSet ELISA Kit | R&D Systems | Cat#DY4517 | |
Commercial assay or kit | Mouse S100a9 DuoSet ELISA Kit | R&D Systems | Cat#DY2065 | |
Commercial assay or kit | PowerUp SYBR Green Master Mix for qPCR | Applied Biosystems (Thermo Fisher) | Cat#A25742 | |
Commercial assay or kit | SuperScript VILO cDNA Synthesis Kit | Thermo Fisher | Cat#11766500 | |
Commercial assay or kit | eBioscience Fixable Viability Dye eFluor 780 | Thermo Fisher | Cat#65-0865-14 | FC (1:1000) |
Chemical compound, drug | fMLP (N-Formyl-Met-Leu-Phe) | Sigma-Aldrich | Cat#F3506 | Neutrophil stimulation: 1 µM |
Chemical compound, drug | PMA (Phorbol 12-myristate 13-acetate) | Sigma-Aldrich | Cat#79346 | Neutrophil stimulation: 100 nM |
Chemical compound, drug | Cytochalasin D | Sigma-Aldrich | Cat#C8273 | Incubated cells at 10 µM |
Chemical compound, drug | SB328437 [ | Tocris Bioscience | Cat#3650 | CCR3 antagonist (10 µM) |
Chemical compound, drug | BI-6901 ( | Gift from Boehringer-Ingelheim Pharma GmbH & Co KG | CCR10 antagonist (20 µM) | |
Chemical compound, drug | Xylazine | VetOne | Cat#RX-0065 | Used for temporary anesthesia: 10 mg/kg, i.p. |
Chemical compound, drug | Ketamine | Zoetis | Cat#000680 | Used for temporary anesthesia: 100 mg/kg, i.p |
Chemical compound, drug | Nalidixic acid sodium salt | Fisher Scientific | Cat#AAJ6355014 | 50 µg/ml for selection |
Chemical compound, drug | Streptomycin sulfate | Fisher Scientific | Cat#5711 | For oral gavage (20 mg/mouse) |
Software, algorithm | GraphPad Prism 10.0 | GraphPad Software | RRID:SCR_002798 | |
Software, algorithm | FlowJo 10.8.1 | BD Biosciences | RRID:SCR_008520 | |
Software, algorithm | QuantStudio 5 Reat-Time PCR System | Thermo Fisher Scientific | RRID:SCR_020240 | |
Software, algorithm | QuPath Analysis Software | QuPath (PMID:29203879) | RRID:SCR_018257 | |
Other | DMSO | Millipore Sigma | Cat#EM-MX1458-6 | Used at 0.1% for vehicle for cytochalasin D during in vitro infection assays described in the Materials and methods |
Other | 2′,7′-Dichlorodihydrofluorescein diacetate | Invitrogen | Cat#D399 | Used at 25 µM for incubation of neutrophils for detection of ROS production by neutrophils, as described in the Materials and methods |
Other | TRI Reagent | Sigma-Aldrich | Cat#T9424 | Used for RNA isolation from tissues, described in Materials and methods section ‘RNA extraction and qPCR’ |
Other | SlowFade Gold Antifade Mountant | Invitrogen | Cat#36936 | Used for staining and mounting immunoflourescent lung sections, described in Materials and methods section ‘Immunofluorescence’ |
Other | APC/Cy7 Streptavidin | BioLegend | Cat#405208 | For tagging biotin-conjugated anti-human myeloperoxidase; FC (1:1000) |
Other | OneComp eBeads | Thermo Fisher | Cat#01-1111-42 | Added to cells at 5 × 105 beads per 1 × 106 cells, as described in the Materials and methods section ‘In vitro neutrophil stimulation’ |
Other | Collagenase, Type VIII | Sigma-Aldrich | Cat#C2139 | For tissue digestion, as described in the Materials and methods: 1 mg/ml |
Other | Liberase | Sigma-Aldrich | Cat#5401020001 | For tissue digestion, as described in the Materials and methods: 20 µg/ml |
Other | DNase I | Sigma-Aldrich | Cat#DN25 | For tissue digestion, as described in the Materials and methods: 0.25 mg/ml |
Other | Helix NP Green | BioLegend | Cat#425303 | For staining neutrophil DNA, as described in the Materials and methods. FC: 10 nM; immuno-fluorescence: 5 µM |
Other | LB Broth, Miller | Fisher Scientific | Cat#DF0446-17-3 | Used for routine culturing of |
Other | LB agar, Miller | Fisher Scientific | Cat#DF0445-17-4 | Used for growth and enumeration of |
Other | Mueller-Hinton Broth | Fisher Scientific | Cat#DF0757-17-6 | Used for routine culturing of |
Other | DPBS | Gibco | Cat#14190250 | Used for washing or resuspension of various cells and bacteria, as described throughout the Materials and methods section |
Other | cOmplete, Mini, EDTA-free Protease Inhibitor Cocktail | Sigma-Aldrich | Cat#4693159001 | Used for fecal protease inhibition as described in the Materials and methods |
Other | Fetal bovine serum (FBS), heat-inactivated | Gibco | Cat#A3840001 | Used for general cell preservation and assays as described in the Materials and methods |
Other | Antibiotic–antimycotic | Gibco | Cat#15-240-062 | Used for general tissue cell preservation as described in the Materials and methods |
Other | RPMI 1640 Medium, with | Gibco | Cat#11875-119 | Used for general tissue cell preservation and assays as described in the Materials and methods |
Other | RPMI 1640 Medium, no glutamine, no phenol red | Gibco | Cat#32404014 | Used for H2DCFDA ROS assays as described in the Materials and methods |
Other | IMDM | Gibco | Cat#12440061 | Used for gut tissue cell isolation as described in the Materials and methods |
Other | Hank’s Balanced Salt Solution | Fisher Scientific | Cat#MT21021CV | Used for gut tissue cell isolation as described in the Materials and methods |
Other | HEPES | Gibco | Cat#15630080 | Used for general tissue cell preservation and assays as described in the Materials and methods |
Other | EDTA | Fisher Scientific | Cat#S311-500 | Used for collection of mouse blood, and for lung and gut tissue cells isolation as described in Materials and methods section ‘Cell extraction and analysis’ |
Other | Bovine serum albumin (BSA) | Fisher Scientific | Cat#BP9703100 | Added to various media for the purpose of blocking non-specific interactions, as described in the Materials and methods sections ‘Cell extraction and analysis’ and ‘Chemotaxis assay’ |
Generation and breeding of
The first colony of
F: 5′-
All mouse experiments were conducted with cohoused wild-type and
For the
For the murine pneumonia model,
CCL28 ELISA
Fresh fecal and blood samples were collected at 4 days post-infection from wild-type mice for quantification of CCL28. Fecal pellets were weighed, resuspended in 1 ml of sterile PBS containing a protease inhibitor cocktail (Roche), and incubated at room temperature shaking for 30 min. Whole-blood samples were collected by cardiac puncture and allowed to clot at room temperature for 30 min. Samples were centrifuged at 9391 ×
Cell extraction and analysis
For the
In vitro neutrophil stimulation
Neutrophils were obtained from the bone marrow of C57BL/6 wild-type mice using the EasySep Mouse Neutrophil Enrichment Kit (STEMCELL), following the manufacturer’s instructions. After enrichment, 1 × 106 neutrophils were seeded per well in a round-bottom 96-well plate with Roswell Park Memorial Institute (RPMI) media supplemented with 10% FBS, 1% antibiotic/antimycotic mix, and 1 mM HEPES (Invitrogen). For stimulation, cells were incubated with LPS-B5 (100 ng/ml, Invivogen), fMLP (1 µM, Sigma-Aldrich), PMA (100 nM, Sigma-Aldrich), and the following concentrations of recombinant mouse cytokines in combination: TNFɑ (100 ng/ml), IFNγ (500 U/ml), and GM-CSF (10 ng/ml), all from BioLegend. For indicated experiments, polystyrene beads (Thermo Fisher) were added to neutrophils at a 1:1 (vol:vol) ratio (MOI = 0.5). Cells were incubated with stimuli for 4 hr at 37°C and 5% CO2. After incubation, cells were recovered by centrifugation, washed with PBS, and processed for flow cytometry as described above.
Chemotaxis assay
Enriched neutrophils from the bone marrow of wild-type mice were stimulated with a cocktail of mouse recombinant cytokines (TNFɑ, IFNγ, GM-CSF), as described above, to induce receptor expression. After stimulation, cells were washed twice with PBS, resuspended in RPMI media supplemented with 0.1% BSA (wt/vol) to a final concentration of 1 × 107 cells/ml, and 100 μl of the cell suspension were placed in the upper compartment of a Transwell chamber (3.0 μm pore size; Corning Costar). 50 nM of recombinant mouse CCL28, CCL11 (R&D Systems), or CXCL1 (Peprotech) were placed into the lower compartment of a Transwell chamber. The Transwell plate was then incubated for 2 hr at 37°C. The number of cells that migrated to the lower compartment was determined by flow cytometry. The neutrophil chemotaxis index was calculated by dividing the number of cells that migrated in the presence of a chemokine by the number of cells that migrated in control chambers without chemokine stimulation.
Neutrophil in vitro infection and bacterial killing assays
Bone marrow neutrophils were obtained from mice as described above.
ROS production
Neutrophils were obtained from the bone marrow of C57BL/6 wild-type mice using the EasySep Mouse Neutrophil Enrichment Kit (STEMCELL Technologies), following the manufacturer’s instructions. After enrichment, 2.5 × 106 cells/ml were resuspended in phenol red-free RPMI media (Gibco) supplemented with 10% FBS (Gibco), and 1 mM HEPES (Invitrogen). The cells were incubated in presence of 2′,7′-dichlorodihydrofluorescein diacetate (H2DCFDA, 25 µM) (Invitrogen), protected from light, for 30 min at 37°C and 5% of CO2, as previously described (Cao et al., 2021). After incubation with H2DCFDA, neutrophils were infected with STm as described above, then incubated for 4 hr with mouse recombinant CCL28 (50 nM), anti-mouse CCR3 antibody (5 µg/1 × 106 cells, clone 83103), anti-mouse CCR10 antibody (5 µg/1 × 106 cells, clone 248918), or anti-rat IgG2A (5 µg/1 × 106 cells, clone 54447), all from R&D Systems. Neutrophils were analyzed by flow cytometry for DCF fluorescence (Ex: 492–495 nm, Em: 517–527 nm) to determine intracellular ROS production using a BD FACSCanto II flow cytometer, and data were analyzed using the FlowJo v10 software.
NETs production
Whole-blood samples were collected from healthy donors recruited at a tertiary care center in Mexico City (Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán). Healthy donors signed an informed consent form before inclusion in the study, and the protocol was approved by the Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán ethics and research committees (Ref. 3341) in compliance with the Helsinki declaration. Neutrophils were obtained from peripheral blood of healthy voluntary donors using the EasySep Direct Human Neutrophil Isolation Kit (STEMCELL Technologies), following the manufacturer’s instructions. In parallel, platelets from human peripheral blood were isolated as described (Du et al., 2018). Briefly, whole blood was centrifuged at 200 ×
Growth of bacteria in media supplemented with recombinant chemokines
RNA extraction and qPCR
Total RNA was extracted from mouse cecal or lung tissue using Tri-Reagent (Molecular Research Center). Reverse transcription of 1 μg of total RNA was performed using the SuperScript VILO cDNA Synthesis kit (Thermo Fisher Scientific). Quantitative real-time PCR for the expression of
Histopathology
Cecal and lung tissue samples collected at necropsy were fixed in 10% buffered formalin for 24–48 hr, then transferred to 70% ethanol for storage. Tissues were embedded in paraffin according to standard procedures and sectioned at 5 μm. Pathology scores of cecal and lung samples were determined by blinded examinations of hematoxylin and eosin-stained sections. Each cecal section was evaluated using a semiquantitative score as described previously (Moschen et al., 2016). Lung inflammation was assessed by a multiparametric scoring based on previous work (Lammers et al., 2012).
Immunofluorescence
Deparaffinized lung sections were stained with a purified rat anti-mouse Ly6G antibody (clone 1A8, BioLegend) according to standard immunohistochemical procedures. Ly6G+ cells were visualized by a goat anti-rat secondary antibody (Invitrogen). Cell nuclei were stained with DAPI in SlowFade Gold Antifade Mountant (Invitrogen). Slides were scanned on a Zeiss Axio Scan.Z1 slide scanner and whole lung scans were evaluated with QuPath analysis software (Bankhead et al., 2017). Ly6G+ cells per mouse were quantified by averaging the neutrophil numbers of three consecutive high-power fields in regions with moderate to severe inflammation.
Statistical analysis
Statistical analysis was performed with GraphPad Prism 10. CFU data from in vivo infection experiments, percentage of CCR3+ or CCR10+ neutrophils in vivo and in vitro, and data from neutrophil functional assays were transformed to Log10 and passed a normal distribution test before running statistical analyses. Data on cytokine secretion, qPCR data, and relative cell abundances within tissues were compared by Mann–Whitney
You have requested "on-the-fly" machine translation of selected content from our databases. This functionality is provided solely for your convenience and is in no way intended to replace human translation. Show full disclaimer
Neither ProQuest nor its licensors make any representations or warranties with respect to the translations. The translations are automatically generated "AS IS" and "AS AVAILABLE" and are not retained in our systems. PROQUEST AND ITS LICENSORS SPECIFICALLY DISCLAIM ANY AND ALL EXPRESS OR IMPLIED WARRANTIES, INCLUDING WITHOUT LIMITATION, ANY WARRANTIES FOR AVAILABILITY, ACCURACY, TIMELINESS, COMPLETENESS, NON-INFRINGMENT, MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE. Your use of the translations is subject to all use restrictions contained in your Electronic Products License Agreement and by using the translation functionality you agree to forgo any and all claims against ProQuest or its licensors for your use of the translation functionality and any output derived there from. Hide full disclaimer
© 2024, Walker, Perez-Lopez et al. This work is published under https://creativecommons.org/licenses/by/4.0/ (the “License”). Notwithstanding the ProQuest Terms and Conditions, you may use this content in accordance with the terms of the License.
Abstract
The chemokine CCL28 is highly expressed in mucosal tissues, but its role during infection is not well understood. Here, we show that CCL28 promotes neutrophil accumulation in the gut of mice infected with
You have requested "on-the-fly" machine translation of selected content from our databases. This functionality is provided solely for your convenience and is in no way intended to replace human translation. Show full disclaimer
Neither ProQuest nor its licensors make any representations or warranties with respect to the translations. The translations are automatically generated "AS IS" and "AS AVAILABLE" and are not retained in our systems. PROQUEST AND ITS LICENSORS SPECIFICALLY DISCLAIM ANY AND ALL EXPRESS OR IMPLIED WARRANTIES, INCLUDING WITHOUT LIMITATION, ANY WARRANTIES FOR AVAILABILITY, ACCURACY, TIMELINESS, COMPLETENESS, NON-INFRINGMENT, MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE. Your use of the translations is subject to all use restrictions contained in your Electronic Products License Agreement and by using the translation functionality you agree to forgo any and all claims against ProQuest or its licensors for your use of the translation functionality and any output derived there from. Hide full disclaimer