Skip to main content
PLOS ONE logoLink to PLOS ONE
. 2021 May 12;16(5):e0247895. doi: 10.1371/journal.pone.0247895

The IL-10GFP (VeRT-X) mouse strain is not suitable for the detection of IL-10 production by granulocytes during lung inflammation

Müge Özkan 1, Yusuf Cem Eskiocak 2, Gerhard Wingender 2,3,*
Editor: Lynette Beattie4
PMCID: PMC8115804  PMID: 33979348

Abstract

The clear and unequivocal identification of immune effector functions is essential to understand immune responses. The cytokine IL-10 is a critical immune regulator and was shown, for example, to limit pathology during various lung diseases. However, the clear identification of IL-10-producing cells is challenging and, therefore, reporter mouse lines were developed to facilitate their detection. Several such reporter lines utilize GFP, including the IL-10GFP (VeRT-X) reporter strain studied here. In line with previous reports, we found that this IL-10GFP line faithfully reports on the IL-10 production of lymphoid cells. However, we show that the IL-10GFP reporter is not suitable to analyse IL-10 production of myeloid cells during inflammation. During inflammation, the autofluorescence of myeloid cells increased to an extent that entirely masked the IL-10-specific GFP-signal. Our data illustrate a general and important technical caveat using GFP-reporter lines for the analysis of myeloid cells and suggest that previous reports on effector functions of myeloid cells using such GFP-based reporters might require re-evaluation.

Introduction

Regulatory cytokines are important players during pulmonary inflammation to limit immunopathology without hampering effective pathogen clearance [1]. The regulatory cytokine IL-10 is involved in various lung diseases, like asthma, allergic airway disease (AAD), chronic obstructive pulmonary disease (COPD), and pulmonary infections [1]. IL-10 producing cells in the lung; FoxP3+ and FoxP3- regulatory T cells [1], CD8+ T cells [2], B cells, alveolar macrophages (AM) [3], interstitial macrophages (IM) [4], airway and alveolar epithelial cells [1], and airway-associated dendritic cells (DCs) [1] were shown to limit disease pathology. However, due to the low percentage of IL-10+ cells within most cell populations and due to the low intensity of the flow cytometric staining of intracellular IL-10 the clear identification of IL-10 producing cells is challenging [5]. Therefore, various reporter mouse lines were developed to avoid the need for intracellular cytokine staining to identify IL-10 producing cells [6]. The IL-10GFP (VeRT-X) strain studied here, expresses an (IRES)-enhanced green fluorescent protein (eGFP) fusion protein downstream of the exon 5 of the il10 gene [7], and was reported to enable the identification of IL-10+ lymphoid but also myeloid cells [7]. Several myeloid cells possess an autofluorescence around 525 nm [8], which coincides with the emission maxima of GFP around 530 nm [9]. However, the impact of this myeloid autofluorescence on the detection of GFP-reporter signals has not been fully clarified. Here, using an LPS-induced lung inflammation model, we demonstrate that the autofluorescence of myeloid cells conceals the IL10GFP-specific signal. This was mainly due to a large increase in the myeloid autofluorescence during the inflammation. These data demonstrate that not all GFP-reporter mouse strains are suitable to analyse effector functions of myeloid cells during inflammation.

Results and discussion

Neutrophils were suggested to be the primary source of IL-10 in the lung following Klebsiella pneumoniae ST258 infection [10]. Furthermore, long-term exposure to house-dust mite (HDM) extracts suppressed allergic airway diseases via the IL-10 production by presumably FoxP3+ T cells and alveolar macrophages [3]. In both studies, the ‘shift’ in the GFP-signal in the experimental IL-10GFP group was compared to either the uninfected IL-10GFP group or the naïve C57BL/6 group as control. When we analysed the IL-10 production during lung inflammation, we noticed that the background signal in the GFP-channel (505 nm—550 nm) of leukocytes from the lung, spleen, or mediastinal lymph node (mLN) was comparable in non-inflamed controls of either C57BL/6 (WT) or IL-10GFP mice (Fig 1A). Upon LPS-induced lung inflammation, the GFP-signal clearly increased, in particular in the lungs (Fig 1B). Nevertheless, the GFP-signal from WT and IL-10GFP mouse-derived cells remained comparable for the percentage (Fig 1A and 1B) or the mean fluorescence intensity (MFI, Fig 1C), irrespective of the organ analysed and the inflammatory condition. These data suggest that the GFP-signal detected in the IL-10GFP—derived cells was actually a background signal.

Fig 1. The myeloid-derived GFP signal is comparable between IL10GFP and C57BL/6 mice and increases during lung inflammation.

Fig 1

C57BL/6 and IL-10GFP mice were challenged three times (d0, d1, d2) with either PBS or 10 μg LPS per mouse via aspiration. 16–18 hours after the last administration, the single cell suspension from lungs, spleens, and mLNs were stained for CD4+ T cells (CD45+ CD45R/B220- CD3ε+ CD4+ CD8α-), CD8+ T cells (CD45+ CD45R/B220- CD3ε+ CD4- CD8α+), macrophages (CD45+ CD45R/B220- CD3ε- Siglec-F- F4/80+), Siglec-F+ cells (eosinophils, alveolar macrophages; CD45+ CD45R/B220- CD3ε- Siglec-F+ F4/80-/+), neutrophils (CD45+ CD45R/B220- CD3ε- Siglec-F- F4/80- Ly6G+ CD11b+), monocytes (CD45+ CD45R/B220- CD3ε- Siglec-F- F4/80- Ly6G- Ly6C+ CD11b+), and ILCs (CD45+ CD45R/B220- CD3ε- Siglec-F- F4/80- Ly6G- CD90.2+ CD127lo/-). The expression of IL-10 was measured either by GFP+ or by intracellular IL-10 as indicated. (A, B) The frequency of GFP+ cells (live CD45+) in the lungs of (A) PBS or (B) LPS challenged C57BL/6 (red) and IL-10GFP (green) mice are shown. The graph shows combined data from three independent experiments (PBS: n = 9 mice/group, LPS: n = 13–15 mice/group in total). (C) Representative histogram showing the shift in the GFP-signal (505 nm—550 nm) of live CD45+ lung cells upon LPS administration in C57BL/6 and IL-10GFP mice strains.

To clarify which cell types could produce IL-10, we directly compared the IL-10 signal derived from the GFP-signal or from intracellular cytokine staining (ICCS) for CD4+ and CD8+ T cells, neutrophils, macrophages, Siglec-F+ cells (eosinophils, alveolar macrophages), monocytes, and ILCs. Cells from the lungs, spleens, and mLN of PBS (control) and LPS-challenged C57BL/6 and IL-10GFP mice were analysed (S1 Fig). Although intracellular IL-10 staining is challenging, we obtained a clear IL-10 signal with the commercial antibody (S2 Fig). With this side-by-side comparison, we found that the IL-10-signal derived from GFP or ICCS correlated well for CD4+ (Fig 2A) and CD8+ T cells (Fig 2B). These data indicate that the IL-10GFP—signal faithfully reports on the IL-10 production of lymphoid cells. However, the GFP-signal from neutrophils (Fig 2C), macrophages (Fig 2D), and Siglec-F+ (Fig 2E) cells was significantly higher than the ICCS-derived signal. Representative dot-plots for all cell types and organs are provided in S3 Fig. Furthermore, the additional staining with a secondary αGFP-AF488-antibody was not able to improve the specificity of the IL-10-signal (S4 Fig). Interestingly, the GFP-signal of monocytes (Fig 2F) and ILCs (Fig 2G) was substantially higher only in the lung tissues, indicating that the increase in the GFP-channel autofluorescence is organ-specific for some cell types.

Fig 2. The myeloid-derived IL-10GFP signal is masked by autofluorescence in granulocytes.

Fig 2

C57BL/6 and IL-10GFP mice were challenged three times (d0, d1, d2) with either PBS or 10 μg LPS per mouse via aspiration. 16–18 hours after the last administration, the single cell suspension from lungs, spleens, and mLNs were stained for CD4+ T cells (CD45+ CD45R/B220- CD3ε+ CD4+ CD8α-), CD8+ T cells (CD45+ CD45R/B220- CD3ε+ CD4- CD8α+), macrophages (CD45+ CD45R/B220- CD3ε- Siglec-F- F4/80+), Siglec-F+ cells (eosinophils, alveolar macrophages; CD45+ CD45R/B220- CD3ε- Siglec-F+ F4/80-/+), neutrophils (CD45+ CD45R/B220- CD3ε- Siglec-F- F4/80- Ly6G+ CD11b+), monocytes (CD45+ CD45R/B220- CD3ε- Siglec-F- F4/80- Ly6G- Ly6C+ CD11b+), and ILCs (CD45+ CD45R/B220- CD3ε- Siglec-F- F4/80- Ly6G- CD90.2+ CD127lo/-). The expression of IL-10 was measured either by GFP+ or by intracellular IL-10 as indicated. (A-G) Comparison of the IL-10 signal derived from intracellular cytokine staining (ICCS) or from the GFP-signal (IL10GFP) of IL10GFP mice in (D) CD4+ T cells, (E) CD8+ T cells, (F) neutrophils, (G) macrophages, (H) Siglec-F+ cells (eosinophils, alveolar macrophages), (I) monocytes, and (J) ILCs from indicated organs. The graph shows combined data from two independent experiments (PBS: n = 6 mice/group, LPS: n = 7–9 mice/group in total).

An overlay of the GFP-signals of CD4+ T cells, CD8+ T cells, neutrophils, macrophages, Siglec-F+ cells, monocytes, and ILCs from control C57BL/6 lung (Fig 3A) and spleen (Fig 3B) indicated that the background GFP-signal was mainly derived from granulocytes. According to the background GFP-signal, cell types ranked as Siglec-F > macrophage > neutrophil > monocyte > ILC. A similar increase in the GFP-channel autofluorescence upon inflammation was noted when alveolar macrophages were analysed (Fig 4).

Fig 3. The cell-type-specific autofluorescence in C57BL/6 mice in control conditions.

Fig 3

Representative histograms from the (A) lung and the (B) spleen of PBS challenged C57BL/6 mice, demonstrating the increase in autofluorescence in the GFP-channel for myeloid cells. The graphs show representative data from three independent experiments (PBS: n = 9 mice/group, LPS: n = 13–15 mice/group in total).

Fig 4. The autofluorescence of alveolar macrophages increases during lung inflammation.

Fig 4

C57BL/6 and IL-10GFP mice were subjected to three daily administrations (d0, d1, d2) of 10 μg LPS per mouse via aspiration. 16–18 hours after the last administration, the single-cell suspensions from the lungs were analysed. Representative histogram showing the GFP-signal from alveolar macrophages (CD45+ CD45R/B220- CD3ε- Siglec-F+ F4/80+ CD11b-) from C57BL/6 and IL10GFP mice as indicated. The data shown are representative of three independent experiments (PBS: n = 9 mice/group, LPS: n = 13–15 mice/group in total).

In conclusion, our data indicate that the IL-10GFP (VeRT-X) reporter strain is not suitable to analyse IL-10 production of myeloid cells during inflammation, due to the strong increase in the autofluorescence, which masks the IL-10-specific GFP-signal. Although we only analysed lung, spleen, and mLN of the VeRT-X IL-10GFP reporter strain, it appears likely that this problem will also be relevant to other organs. Importantly, the strong increase of the granulocytic autofluorescence in the GFP-channels was independent of the strain analysed. Therefore, it is expected that other fluorescent reporter lines that utilize the GFP-channel would face similar problems at distinguishing the reporter-specific signal from the autofluorescence signal when analysing granulocytes. Our data indicate that granulocytes are the source of the background GFP-signal, in line with previous publications reporting high levels of autofluorescence [11], although other sources, like collagen deposition [12,13], cannot be excluded. This large increase in false-positive GFP-signals for myeloid cells during inflammation could have been missed previously due to a focus on lymphoid cells or due to a lack of the WT-treated controls. Our data illustrate a general and important technical caveat using GFP-reporter lines for the analysis of myeloid cells and suggest that previous reports on effector functions of myeloid cells using such GFP-based reporters might require re-evaluation.

Material and methods

Mice

C57BL/6J mice and IL-10GFP (also known as Vert-X) B6(Cg)-Il10tm1.1Karp/J mice were originally obtained from Jackson Laboratory (Bar Harbor, USA). All mice were housed in the vivarium of the Izmir Biomedicine and Genome Center (IBG, Izmir, Turkey) in accordance with the respective institutional animal care committee guidelines. All mouse experiments were performed with prior approval by the institutional ethic committee (‘Izmir Biomedicine and Genome Center’s Ethical Committee on Animal Experimentation’), in accordance with national laws and policies. All the methods were carried out in accordance with the approved guidelines and regulations.

Reagents and monoclonal antibodies

Monoclonal antibodies against the following mouse antigens were used in this study: CD3ε (145.2C11), CD4 (RM4-5), CD8α (53–6.7), CD11b (M1/70), CD11c (N418), CD45 (30-F11), CD45R/B220 (RA3-6B2), CD90.2 (30-H12), CD127 (A7R34), CD170/Siglec-F (E50-2440), F4/80 (BM8), IL-10 (JESS-16E3), Ly6C (HK1.4), Ly6G (1A8). Antibodies were purchased from BD Biosciences (San Diego, USA), BioLegend (San Diego, USA), or ThermoFisher Scientific (Carlsbad, USA). Antibodies were conjugated to Pacific Blue, Brilliant Violet 421, V500, Brilliant Violet 510, Brilliant Violet 570, Brilliant Violet 650, Brilliant Violet 711, Brilliant Violet 785, Brilliant Violet 786, FITC, Alexa Fluor 488, PerCP-Cy5.5, PerCP-eFluor 710, PE, PE-CF594, PE-Cy7, APC, Alexa Fluor 647, Alexa Fluor 700, APC-Cy7, or APC-Fire750. Details on the antibody used in this study are given in the S1 Table. Anti-mouse CD16/32 antibody (2.4G2) and Zombie UV Dead Cell Staining Kit were obtained from Tonbo Biosciences (San Diego, USA) and from BioLegend, respectively. Unconjugated mouse and rat IgG antibodies were purchased from Jackson ImmunoResearch (West Grove, USA).

LPS-induced lung inflammation

Lung inflammation was induced by three daily administrations (d0, d1, d2) with 10 μg LPS (#L6386, Sigma-Aldrich, St. Louis, USA) per mouse via pharyngeal/laryngeal aspiration. 16–18 hours after the last challenge, the mice were sacrificed, and lungs, spleens, and mediastinal lymph nodes were collected.

Cell preparation

Lungs were removed and minced into smaller pieces in a 6-well plate (Greiner, Germany). The digestion mixture, composed of 1 mg/mL collagenase D and 0.1 mg/mL DNase I (both from Roche, Switzerland) in complete RPMI medium (Gibco, USA), was added to the samples and incubated for 45 min at 37°C on a lateral shaker. The lung samples were filtered through 100 μm mesh with PBS, washed twice, and the red blood cells were eliminated by ACK lysis buffer (Lonza, USA). Spleens and mediastinal lymph nodes were homogenized by filtering through a 76 μm mesh with ice-cold PBS (Lonza), washed twice, and red blood cells were eliminated by ACK lysis buffer (Lonza).

In vitro stimulation

Lymphocytes were stimulated in vitro with PMA (50 ng/mL) and ionomycin (1 μg/mL) (both Sigma-Aldrich, St. Louis, MO) for four hours at 37°C in the presence of both Brefeldin A (GolgiPlug) and Monensin (GolgiStop) in complete RPMI medium (RPMI 1640 medium (Life Technologies); supplemented with 10% (v/v) fetal calf serum (FCS), 1% (v/v) Pen-Strep-Glutamine (10.000 U/ml penicillin, 10.000 μg/ml streptomycin, 29.2 mg/ml L-glutamine (Life Technologies)) and 50 μM β-mercaptoethanol (Sigma)). As GolgiPlug and GolgiStop (both BD Biosciences) were used together, half the amount recommended by the manufacturer were used.

Flow cytometry

Flow cytometry was performed as described [14]. In brief, for staining of cell surface molecules, cells were suspended in staining buffer (PBS, 1% BSA, 0.01% NaN3) and stained with fluorochrome-conjugated antibodies (0.1–1 μg/106 cells, or according to the manufacturer’s recommendations) for 15 min in a total volume of 50 μl at 4°C for 30 min. FcεR-blocking antibody αCD16/32 (2.4G2) and unconjugated rat and mouse IgG (Jackson ImmunoResearch) were added to prevent non-specific binding. If biotin-conjugated antibodies were used, cell-bound antibodies were detected with streptavidin conjugates (1:200, or according to the manufacturer’s recommendations) in a second incubation step. Dead cells were labelled with a commercially available Zombie UV Dead Cell Staining Kit (BioLegend). For the analysis of intracellular cytokines, cells were fixed and permeabilized with Cytofix/Cytoperm (BD Biosciences) for 10 min at 37°C. Cells were washed twice and incubated overnight at 4°C with the fluorochrome-conjugated antibodies and unconjugated rat and mouse IgG in Perm/Wash solution (BD Biosciences), which was followed by additional 5 min incubation in Perm/Wash solution without antibodies. For the analysis of the GFP signal, the cells were fixed with freshly prepared 2% formaldehyde solution for 40 minutes on ice as described [15]. This fixation allows the detection of the GFP-signal together with other intracellular proteins [15]. Then, the cells were permeabilized using the BD Perm/Wash solution. Cells were washed twice and incubated overnight at 4°C with the fluorochrome-conjugated antibodies and unconjugated rat and mouse IgG in Perm/Wash solution, which was followed by additional 5 min incubation in Perm/Wash solution without antibodies. Cells were analysed with FACSCanto or LSR-Fortessa (BD Biosciences), and data were processed with CellQuest Pro (BD Biosciences) or Flow Jo (Tree Star) software.

Statistical analysis

Data are presented as mean ± standard error of the mean (SEM). The statistical analysis was performed with GraphPad Prism 7.0 software (GraphPad Software, San Diego, USA). One-way ANOVA followed by Holm-Sidak posthoc test are used to compare p-values regarded as *p≤0.05, **p≤0.01, and ***p≤0.001.

Supporting information

S1 Fig. Gating strategy to identify leukocytes from the lung, spleen, and mLN.

A graphic outline (A) and exemplary graphs (B) are given to illustrate the gating strategy employed to identify CD4+ T cells (CD45+ CD45R/B220- CD3ε+ CD4+ CD8α- CD127lo), CD8+ T cells (CD45+ CD45R/B220- CD3ε+ CD4- CD8α+), macrophages (CD45+ CD45R/B220- CD3ε- Siglec-F- F4/80+), Siglec-F+ cells (eosinophils, alveolar macrophages; CD45+ CD45R/B220- CD3ε- Siglec-F+ F4/80-/+), neutrophils (CD45+ CD45R/B220- CD3ε- Siglec-F- F4/80- Ly6G+ CD11b+), monocytes (CD45+ CD45R/B220- CD3ε- Siglec-F- F4/80- Ly6G- Ly6C+ CD11b+), and ILCs in the inflamed lung.

(TIF)

S2 Fig. Efficiency of intracellular IL-10 cytokine staining.

Representative flow cytometric dot plots showing the IL-10 staining (ICCS) in splenic CD4+ T cells.

(TIF)

S3 Fig. Representative flow cytometry plots for the data presented in Fig 2.

C57BL/6 and IL-10GFP mice were challenged three times (d0, d1, d2) with either PBS or 10 μg LPS per mouse via aspiration. 16–18 hours after the last administration, the single cell suspension from (A) lungs, (B) spleens, and (C) mLNs were stained for CD4+ T cells (CD45+ CD45R/B220- CD3ε+ CD4+ CD8α-), CD8+ T cells (CD45+ CD45R/B220- CD3ε+ CD4- CD8α+), macrophages (CD45+ CD45R/B220- CD3ε- Siglec-F- F4/80+), Siglec-F+ cells (eosinophils, alveolar macrophages; CD45+ CD45R/B220- CD3ε- Siglec-F+ F4/80-/+), neutrophils (CD45+ CD45R/B220- CD3ε- Siglec-F- F4/80- Ly6G+ CD11b+), monocytes (CD45+ CD45R/B220- CD3ε- Siglec-F- F4/80- Ly6G- Ly6C+ CD11b+), and ILCs (CD45+ CD45R/B220- CD3ε- Siglec-F- F4/80- Ly6G- CD90.2+ CD127lo/-). The expression of IL-10 was measured either by intracellular IL-10 staining (left panel) or GFP—expression (right panel).

(TIF)

S4 Fig. The staining with a secondary αGFP-AF488 antibody does not improve the resolution of the myeloid IL10GFP signal.

Representative histograms from the spleen of PBS (left) or LPS (right) challenged C57BL/6 and IL-10GFP mouse, demonstrating the overall GFP-signal detected with or without labelling with secondary αGFP-AF488.

(TIF)

S1 Table

(TIF)

Abbreviations

AAD

allergic airway disease

AM

alveolar macrophage

COPD

chronic obstructive pulmonary disease

DC

dendritic cell

HDM

house dust mite

ICCS

intracellular cytokine staining

ILC

innate lymphoid cell

IM

interstitial macrophages

GFP

green fluorescent protein

LPS

lipopolysaccharide

mLN

mediastinal lymph node

Data Availability

All relevant data are within the manuscript and its Supporting Information files.

Funding Statement

This work was funded by grants from the Scientific and Technological Research Council of Turkey (TUBITAK, www.tubitak.gov.tr; #116Z272, GW), the Dokuz Eylul University (Izmir, Turkey; www.deu.edu.tr; #2020.KB.SAG.031, GW), and the European Molecular Biology Organization (EMBO, www.embo.org; IG3073; GW). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

References

  • 1.Branchett WJ, Lloyd CM. Regulatory cytokine function in the respiratory tract. Mucosal Immunol [Internet]. 2019. Mar 15;12(3):589–600. Available from: 10.1038/s41385-019-0158-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Sun J, Madan R, Karp CL, Braciale TJ. Effector T cells control lung inflammation during acute influenza virus infection by producing IL-10. Nat Med. 2009;15(3):277–84. 10.1038/nm.1929 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Bracken SJ, Adami AJ, Szczepanek SM, Ehsan M, Natarajan P, Guernsey LA, et al. Long-Term Exposure to House Dust Mite Leads to the Suppression of Allergic Airway Disease Despite Persistent Lung Inflammation. Int Arch Allergy Immunol. 2015;166(4):243–58. 10.1159/000381058 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Kawano H, Kayama H, Nakama T, Hashimoto T, Umemoto E, Takeda K. IL-10-producing lung interstitial macrophages prevent neutrophilic asthma. Int Immunol. 2016;28(10):489–501. 10.1093/intimm/dxw012 [DOI] [PubMed] [Google Scholar]
  • 5.Sag D, Özkan M, Kronenberg M, Wingender G. Improved Detection of Cytokines Produced by Invariant NKT Cells. Sci Rep. 2017;7(1). 10.1038/s41598-017-16832-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Bouabe H. Cytokine Reporter Mice: The Special Case of IL-10. Scand J Immunol [Internet]. 2012. Jun;75(6):553–67. Available from: http://doi.wiley.com/10.1111/j.1365-3083.2012.02695.x. [DOI] [PubMed] [Google Scholar]
  • 7.Madan R, Demircik F, Surianarayanan S, Allen JL, Divanovic S, Trompette A, et al. Nonredundant Roles for B Cell-Derived IL-10 in Immune Counter-Regulation. J Immunol. 2009;183(4):2312–20. 10.4049/jimmunol.0900185 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Mitchell AJ, Pradel LC, Chasson L, Van Rooijen N, Grau GE, Hunt NH, et al. Technical Advance: Autofluorescence as a tool for myeloid cell analysis. J Leukoc Biol [Internet]. 2010. September;88(3):597–603. Available from: http://doi.wiley.com/10.1189/jlb.0310184. [DOI] [PubMed] [Google Scholar]
  • 9.Telford WG, Hawley T, Subach F, Verkhusha V, Hawley RG. Flow cytometry of fluorescent proteins. Methods [Internet]. 2012;57(3):318–30. Available from: 10.1016/j.ymeth.2012.01.003 [DOI] [PubMed] [Google Scholar]
  • 10.Peñaloza HF, Noguera LP, Ahn D, Vallejos OP, Castellanos RM, Vazquez Y, et al. Interleukin-10 produced by Myeloid-Derived suppressor cells provides protection to Carbapenem-Resistant klebsiella pneumoniae sequence type 258 by enhancing its clearance in the airways. Infect Immun. 2019;87(5). 10.1128/IAI.00665-18 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Tighe RM, Misharin A V., Jakubzick C V., Brinkman R, Curtis JL, Duggan R, et al. Improving the quality and reproducibility of flow cytometry in the lung. Am J Respir Cell Mol Biol. 2019;61(2):150–61. 10.1165/rcmb.2019-0191ST [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Black PN, Ching PST, Beaumont B, Ranasinghe S, Taylor G, Merrilees MJ. Changes in elastic fibres in the small airways and alveoli in COPD. Eur Respir J. 2008;31(5):998–1004. 10.1183/09031936.00017207 [DOI] [PubMed] [Google Scholar]
  • 13.Kretschmer S, Pieper M, Hüttmann G, Bölke T, Wollenberg B, Marsh LM, et al. Autofluorescence multiphoton microscopy for visualization of tissue morphology and cellular dynamics in murine and human airways. Lab Investig. 2016;96(8):918–31. 10.1038/labinvest.2016.69 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Wingender G, Birkholz a. M, Sag D, Farber E, Chitale S, Howell a. R, et al. Selective Conditions Are Required for the Induction of Invariant NKT Cell Hyporesponsiveness by Antigenic Stimulation. J Immunol [Internet]. 2015;195(8):3838–48. Available from: http://www.jimmunol.org/cgi/doi/10.4049/jimmunol.1500203. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Heinen P, Wanke F, Moos S, Attig S, Waisman A, Kurschus FC, et al. Improved Method to Retain Cytosolic Reporter Protein Fluorescence While Staining for Nuclear Proteins. 2014;(3). 10.1002/cyto.a.22451 [DOI] [PubMed] [Google Scholar]

Decision Letter 0

Lynette Beattie

26 Mar 2021

PONE-D-21-05124

The IL-10GFP (VeRT-X) mouse strain is not suitable for the detection of IL-10 production by granulocytes during lung inflammation

PLOS ONE

Dear Dr. Wingender,

Thank you for submitting your manuscript to PLOS ONE. After careful consideration, we feel that it has merit but does not fully meet PLOS ONE’s publication criteria as it currently stands. Therefore, we invite you to submit a revised version of the manuscript that addresses the points raised during the review process.

Please address the comments of both reviewers, as outlined below, including addition of the additional FACS plots and data as requested.

Please submit your revised manuscript by May 10 2021 11:59PM. If you will need more time than this to complete your revisions, please reply to this message or contact the journal office at plosone@plos.org. When you're ready to submit your revision, log on to https://www.editorialmanager.com/pone/ and select the 'Submissions Needing Revision' folder to locate your manuscript file.

Please include the following items when submitting your revised manuscript:

  • A rebuttal letter that responds to each point raised by the academic editor and reviewer(s). You should upload this letter as a separate file labeled 'Response to Reviewers'.

  • A marked-up copy of your manuscript that highlights changes made to the original version. You should upload this as a separate file labeled 'Revised Manuscript with Track Changes'.

  • An unmarked version of your revised paper without tracked changes. You should upload this as a separate file labeled 'Manuscript'.

If you would like to make changes to your financial disclosure, please include your updated statement in your cover letter. Guidelines for resubmitting your figure files are available below the reviewer comments at the end of this letter.

If applicable, we recommend that you deposit your laboratory protocols in protocols.io to enhance the reproducibility of your results. Protocols.io assigns your protocol its own identifier (DOI) so that it can be cited independently in the future. For instructions see: http://journals.plos.org/plosone/s/submission-guidelines#loc-laboratory-protocols. Additionally, PLOS ONE offers an option for publishing peer-reviewed Lab Protocol articles, which describe protocols hosted on protocols.io. Read more information on sharing protocols at https://plos.org/protocols?utm_medium=editorial-email&utm_source=authorletters&utm_campaign=protocols.

We look forward to receiving your revised manuscript.

Kind regards,

Lynette Beattie, PhD

Academic Editor

PLOS ONE

Journal Requirements:

When submitting your revision, we need you to address these additional requirements.

1. Please ensure that your manuscript meets PLOS ONE's style requirements, including those for file naming. The PLOS ONE style templates can be found at

https://journals.plos.org/plosone/s/file?id=wjVg/PLOSOne_formatting_sample_main_body.pdf and

https://journals.plos.org/plosone/s/file?id=ba62/PLOSOne_formatting_sample_title_authors_affiliations.pdf

2. We note that you have included the phrase “data not shown” in your manuscript. Unfortunately, this does not meet our data sharing requirements. PLOS does not permit references to inaccessible data. We require that authors provide all relevant data within the paper, Supporting Information files, or in an acceptable, public repository. Please add a citation to support this phrase or upload the data that corresponds with these findings to a stable repository (such as Figshare or Dryad) and provide and URLs, DOIs, or accession numbers that may be used to access these data. Or, if the data are not a core part of the research being presented in your study, we ask that you remove the phrase that refers to these data.

3. Please upload a new copy of Supporting Information Figure 2 as the detail is not clear.

Please follow the link for more information: https://blogs.plos.org/plos/2019/06/looking-good-tips-for-creating-your-plos-figures-graphics/

4. Please review your reference list to ensure that it is complete and correct. If you have cited papers that have been retracted, please include the rationale for doing so in the manuscript text, or remove these references and replace them with relevant current references. Any changes to the reference list should be mentioned in the rebuttal letter that accompanies your revised manuscript. If you need to cite a retracted article, indicate the article’s retracted status in the References list and also include a citation and full reference for the retraction notice.

Reviewers' comments:

Reviewer's Responses to Questions

Comments to the Author

1. Is the manuscript technically sound, and do the data support the conclusions?

The manuscript must describe a technically sound piece of scientific research with data that supports the conclusions. Experiments must have been conducted rigorously, with appropriate controls, replication, and sample sizes. The conclusions must be drawn appropriately based on the data presented.

Reviewer #1: Yes

Reviewer #2: Yes

**********

2. Has the statistical analysis been performed appropriately and rigorously?

Reviewer #1: Yes

Reviewer #2: Yes

**********

3. Have the authors made all data underlying the findings in their manuscript fully available?

The PLOS Data policy requires authors to make all data underlying the findings described in their manuscript fully available without restriction, with rare exception (please refer to the Data Availability Statement in the manuscript PDF file). The data should be provided as part of the manuscript or its supporting information, or deposited to a public repository. For example, in addition to summary statistics, the data points behind means, medians and variance measures should be available. If there are restrictions on publicly sharing data—e.g. participant privacy or use of data from a third party—those must be specified.

Reviewer #1: Yes

Reviewer #2: Yes

**********

4. Is the manuscript presented in an intelligible fashion and written in standard English?

PLOS ONE does not copyedit accepted manuscripts, so the language in submitted articles must be clear, correct, and unambiguous. Any typographical or grammatical errors should be corrected at revision, so please note any specific errors here.

Reviewer #1: Yes

Reviewer #2: Yes

**********

5. Review Comments to the Author

Please use the space provided to explain your answers to the questions above. You may also include additional comments for the author, including concerns about dual publication, research ethics, or publication ethics. (Please upload your review as an attachment if it exceeds 20,000 characters)

Reviewer #1: Ozkan et al describe a potential caveat of utilising IL-10 reporter mice (VeRT-X) to identify myeloid-derived IL-10 production during an experimental model of LPS-induced lung inflammation.

The manuscript is well written, and the data is presented in a concise manner. The following comments are suggestions which I believe may add clarity to the overall message of the manuscript.

Major comments:

1. Can the authors comment on how the VeRT-X IL-10 reporter line and their results compare to any of the other eight IL-10 reporter lines, described by Bouabe 2012 (https://doi.org/10.1111/j.1365-3083.2012.02695.x)?

2. The original paper describing the VeRT-X mice (Madan et al., 2009) shows IL-10 GFP expression by B cells. Have the authors measured B cell derived IL-10 GFP levels in their model of LPS-induced lung inflammation?

3. It would be helpful to provide representative FACS plots as part of Supplementary Figure 1, to complement the gating strategy.

4. Figure 2 nicely illustrates IL-10 detection by ICCS vs GFP. It would be helpful to show representative FACS plots as part of this figure or as a Supplementary figure.

5. The figure legend for Figure 2 states: “The graph shows combined data from two independent experiments (PBS: n = 6 mice/group. LPS: n=7-9 mice/group)”. Can the authors confirm whether 6-9 mice/group were used in each individual experiment? Or whether the 6-9mice/group refers to the pooled data shown in Figure 2?

Minor comments:

Line 31: please remove ‘s’ from inflammations

Line 49: please change where to were

Line 54: please insert reference 7 after myeloid cells.

Line 54: please check reference 8, since the autofluorescence of myeloid cells was not mentioned in this reference.

Line 90-91: please clarify why data is not shown from WT controls. It might be informative to show this data as a supplementary figure.

Line 103: please remove ‘s’ from inflammations

Line 108: please remove ‘also’

Line 110: please change ‘analysis’ to analysing

Line 114: please remove ‘s’ from inflammations

Line 145: please add ‘d’ to purchase

Line 318: please superscript IL10GFP

Line 321 – 322: please superscript CD45+ CD45R/B220- CD3- Siglec-F+ F4/80+ CD11b- and IL10GFP

Reviewer #2: In this paper, the authors have proposed that IL-10 GFP reporter strain is not an appropriate model to detect IL-10 production by granulocytes. Using C57BL/6 and IL-10 GFP mice and by employing flowcytometry, the authors show that GFP signal seen in granulocytes post LPS challenge is not the actual GFP signal due to IL-10 production. Rather, it is attributable to autofluorescence. I think the authors have used proper controls in the study and results from the study illustrates important technical caveat in using GFP reporter mice for analysing myeloid cells. Overall, the study is good. However, I have few minor suggestions:

1. Line 31: Please change " during inflammations" to during inflammation. Please make this change elsewhere as well.

2. Line 39-40: Please rephrase this sentence as the meaning is not clear.

3. Line 49: Please change "lines where" to lines were.

4. Line 54: Please give reference when you discuss that IL-10 GFP strain was reported to enable identification of IL-10+

myeloid cells.

5. Line 110: Please change"analysis" to analysing.

6. Line 170: Change "where" to were.

7. Line 277-279 and 295-296: Please rephrase these two sentences as they seem incomplete.

8. Line 337-339: I think this sentence " C) Representative histograms from the spleen of PBS-------" is not required as it is a repetition of Supplementary Figure 3 legend.

9. Please check the overall grammar and sentence structure throughout the text.

**********

6. PLOS authors have the option to publish the peer review history of their article (what does this mean?). If published, this will include your full peer review and any attached files.

If you choose “no”, your identity will remain anonymous but your review may still be made public.

Do you want your identity to be public for this peer review? For information about this choice, including consent withdrawal, please see our Privacy Policy.

Reviewer #1: No

Reviewer #2: No

[NOTE: If reviewer comments were submitted as an attachment file, they will be attached to this email and accessible via the submission site. Please log into your account, locate the manuscript record, and check for the action link "View Attachments". If this link does not appear, there are no attachment files.]

While revising your submission, please upload your figure files to the Preflight Analysis and Conversion Engine (PACE) digital diagnostic tool, https://pacev2.apexcovantage.com/. PACE helps ensure that figures meet PLOS requirements. To use PACE, you must first register as a user. Registration is free. Then, login and navigate to the UPLOAD tab, where you will find detailed instructions on how to use the tool. If you encounter any issues or have any questions when using PACE, please email PLOS at figures@plos.org. Please note that Supporting Information files do not need this step.

Decision Letter 1

Lynette Beattie

27 Apr 2021

The IL-10GFP (VeRT-X) mouse strain is not suitable for the detection of IL-10 production by granulocytes during lung inflammation

PONE-D-21-05124R1

Dear Dr. Wingender,

We’re pleased to inform you that your manuscript has been judged scientifically suitable for publication and will be formally accepted for publication once it meets all outstanding technical requirements.

Within one week, you’ll receive an e-mail detailing the required amendments. When these have been addressed, you’ll receive a formal acceptance letter and your manuscript will be scheduled for publication.

An invoice for payment will follow shortly after the formal acceptance. To ensure an efficient process, please log into Editorial Manager at http://www.editorialmanager.com/pone/, click the 'Update My Information' link at the top of the page, and double check that your user information is up-to-date. If you have any billing related questions, please contact our Author Billing department directly at authorbilling@plos.org.

If your institution or institutions have a press office, please notify them about your upcoming paper to help maximize its impact. If they’ll be preparing press materials, please inform our press team as soon as possible -- no later than 48 hours after receiving the formal acceptance. Your manuscript will remain under strict press embargo until 2 pm Eastern Time on the date of publication. For more information, please contact onepress@plos.org.

Kind regards,

Lynette Beattie, PhD

Academic Editor

PLOS ONE

Additional Editor Comments (optional):

Reviewers' comments:

Reviewer's Responses to Questions

Comments to the Author

1. If the authors have adequately addressed your comments raised in a previous round of review and you feel that this manuscript is now acceptable for publication, you may indicate that here to bypass the “Comments to the Author” section, enter your conflict of interest statement in the “Confidential to Editor” section, and submit your "Accept" recommendation.

Reviewer #1: All comments have been addressed

Reviewer #2: All comments have been addressed

**********

2. Is the manuscript technically sound, and do the data support the conclusions?

The manuscript must describe a technically sound piece of scientific research with data that supports the conclusions. Experiments must have been conducted rigorously, with appropriate controls, replication, and sample sizes. The conclusions must be drawn appropriately based on the data presented.

Reviewer #1: Yes

Reviewer #2: Yes

**********

3. Has the statistical analysis been performed appropriately and rigorously?

Reviewer #1: Yes

Reviewer #2: Yes

**********

4. Have the authors made all data underlying the findings in their manuscript fully available?

The PLOS Data policy requires authors to make all data underlying the findings described in their manuscript fully available without restriction, with rare exception (please refer to the Data Availability Statement in the manuscript PDF file). The data should be provided as part of the manuscript or its supporting information, or deposited to a public repository. For example, in addition to summary statistics, the data points behind means, medians and variance measures should be available. If there are restrictions on publicly sharing data—e.g. participant privacy or use of data from a third party—those must be specified.

Reviewer #1: Yes

Reviewer #2: Yes

**********

5. Is the manuscript presented in an intelligible fashion and written in standard English?

PLOS ONE does not copyedit accepted manuscripts, so the language in submitted articles must be clear, correct, and unambiguous. Any typographical or grammatical errors should be corrected at revision, so please note any specific errors here.

Reviewer #1: Yes

Reviewer #2: Yes

**********

6. Review Comments to the Author

Please use the space provided to explain your answers to the questions above. You may also include additional comments for the author, including concerns about dual publication, research ethics, or publication ethics. (Please upload your review as an attachment if it exceeds 20,000 characters)

Reviewer #1: (No Response)

Reviewer #2: (No Response)

**********

7. PLOS authors have the option to publish the peer review history of their article (what does this mean?). If published, this will include your full peer review and any attached files.

If you choose “no”, your identity will remain anonymous but your review may still be made public.

Do you want your identity to be public for this peer review? For information about this choice, including consent withdrawal, please see our Privacy Policy.

Reviewer #1: No

Reviewer #2: No

Acceptance letter

Lynette Beattie

3 May 2021

PONE-D-21-05124R1

The IL-10GFP (VeRT-X) mouse strain is not suitable for the detection of IL-10 production by granulocytes during lung inflammation

Dear Dr. Wingender:

I'm pleased to inform you that your manuscript has been deemed suitable for publication in PLOS ONE. Congratulations! Your manuscript is now with our production department.

If your institution or institutions have a press office, please let them know about your upcoming paper now to help maximize its impact. If they'll be preparing press materials, please inform our press team within the next 48 hours. Your manuscript will remain under strict press embargo until 2 pm Eastern Time on the date of publication. For more information please contact onepress@plos.org.

If we can help with anything else, please email us at plosone@plos.org.

Thank you for submitting your work to PLOS ONE and supporting open access.

Kind regards,

PLOS ONE Editorial Office Staff

on behalf of

Dr. Lynette Beattie

Academic Editor

PLOS ONE

Associated Data

    This section collects any data citations, data availability statements, or supplementary materials included in this article.

    Supplementary Materials

    S1 Fig. Gating strategy to identify leukocytes from the lung, spleen, and mLN.

    A graphic outline (A) and exemplary graphs (B) are given to illustrate the gating strategy employed to identify CD4+ T cells (CD45+ CD45R/B220- CD3ε+ CD4+ CD8α- CD127lo), CD8+ T cells (CD45+ CD45R/B220- CD3ε+ CD4- CD8α+), macrophages (CD45+ CD45R/B220- CD3ε- Siglec-F- F4/80+), Siglec-F+ cells (eosinophils, alveolar macrophages; CD45+ CD45R/B220- CD3ε- Siglec-F+ F4/80-/+), neutrophils (CD45+ CD45R/B220- CD3ε- Siglec-F- F4/80- Ly6G+ CD11b+), monocytes (CD45+ CD45R/B220- CD3ε- Siglec-F- F4/80- Ly6G- Ly6C+ CD11b+), and ILCs in the inflamed lung.

    (TIF)

    S2 Fig. Efficiency of intracellular IL-10 cytokine staining.

    Representative flow cytometric dot plots showing the IL-10 staining (ICCS) in splenic CD4+ T cells.

    (TIF)

    S3 Fig. Representative flow cytometry plots for the data presented in Fig 2.

    C57BL/6 and IL-10GFP mice were challenged three times (d0, d1, d2) with either PBS or 10 μg LPS per mouse via aspiration. 16–18 hours after the last administration, the single cell suspension from (A) lungs, (B) spleens, and (C) mLNs were stained for CD4+ T cells (CD45+ CD45R/B220- CD3ε+ CD4+ CD8α-), CD8+ T cells (CD45+ CD45R/B220- CD3ε+ CD4- CD8α+), macrophages (CD45+ CD45R/B220- CD3ε- Siglec-F- F4/80+), Siglec-F+ cells (eosinophils, alveolar macrophages; CD45+ CD45R/B220- CD3ε- Siglec-F+ F4/80-/+), neutrophils (CD45+ CD45R/B220- CD3ε- Siglec-F- F4/80- Ly6G+ CD11b+), monocytes (CD45+ CD45R/B220- CD3ε- Siglec-F- F4/80- Ly6G- Ly6C+ CD11b+), and ILCs (CD45+ CD45R/B220- CD3ε- Siglec-F- F4/80- Ly6G- CD90.2+ CD127lo/-). The expression of IL-10 was measured either by intracellular IL-10 staining (left panel) or GFP—expression (right panel).

    (TIF)

    S4 Fig. The staining with a secondary αGFP-AF488 antibody does not improve the resolution of the myeloid IL10GFP signal.

    Representative histograms from the spleen of PBS (left) or LPS (right) challenged C57BL/6 and IL-10GFP mouse, demonstrating the overall GFP-signal detected with or without labelling with secondary αGFP-AF488.

    (TIF)

    S1 Table

    (TIF)

    Attachment

    Submitted filename: Point-to-point response_Ozkan et al.pdf

    Data Availability Statement

    All relevant data are within the manuscript and its Supporting Information files.


    Articles from PLoS ONE are provided here courtesy of PLOS

    RESOURCES