SPLUNC1 knockout enhances LPS-induced lung injury by increasing recruitment of CD11b+Gr-1+ cells to the spleen of mice

  • Authors:
    • Han Zhang
    • Xiaoling Li
    • Shan Liao
    • Heran Wang
    • Pan Chen
    • Guangchao Zhu
    • Yanwei Luo
    • Guiyuan Li
    • Yanhong Zhou
  • View Affiliations

  • Published online on: November 1, 2017     https://doi.org/10.3892/or.2017.6063
  • Pages: 358-366
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Short palate, lung and nasal epithelium clone 1 (SPLUNC1) is a tissue-specific gene of nasopharyngeal tissue, and has been recognized as a potential tumor-suppressor gene in nasopharyngeal carcinoma. As a secreted protein, SPLUNC1 plays an important role in innate immunity including antimicrobial and host defense. However, the related immune cells which are regulated by SPLUNC1 remain elusive. In the present study, an acute lung injury (ALI) mouse model was established by administration of lipopolysaccharide (LPS) intraperitoneal injections to wild-type and SPLUNC1-/- mice (5 mg/kg). Pathologic results showed that the SPLUNC1-/- group appeared to have more severe pulmonary damage and infiltrated inflammatory cells compared with the WT group after LPS treatment for 24, 48, 72 and 96 h. The mRNA expression levels of interleukin-6 (IL-6), chemokine (C-C motif) ligand-2 (CCL-2), chemokine (C-C motif) ligand-3 (CCL-3) and chemokine (C-X-C motif) ligand-1 (CXCL-1) in lungs of the SPLUNC1-/- group were higher than these levels in lungs of the WT group at different time points after LPS injection. The percentage of splenic CD11b+Gr-1+ myeloid-derived suppressor cells (MDSCs) in the SPLUNC1-/- mice was higher than this percentage in the WT mice at the time points of 72 and 96 h post LPS injection (P<0.05). These findings demonstrated that SPLUNC1 had a certain protective effect on the LPS-induced ALI mouse model as well as it was found to inhibit the recruitment of MDSCs to the spleen in this model.

Introduction

Acute lung injury (ALI) is a complicated pathogenic process with high mortality due to sepsis, pulmonary infection and shock (1). Lipopolysaccharide (LPS), a component found in the cell membrane of gram-negative bacteria, induces the production of numerous inflammatory cytokines and chemokines and has been shown as a major cause of ALI (2). Intraperitoneal administration of LPS has been widely used to model the pathogenesis of ALI and has yielded reproducible results.

Short palate, lung and nasal epithelium clone 1 (SPLUNC1) is a protein mainly secreted by large airway epithelia and submucosal glands (3,4). Accordingly, we can find this protein in the human oral cavity (5), nasal secretions (6), tracheal aspirates (7) as well as in rat lung (8), suggesting that SPLUNC1 is a highly abundant secretory product of the mammalian respiratory system. Recent studies by our group demonstrated that SPLUNC1 inhibits the overall process of Epstein Barr virus replication and is associated with nasopharyngeal carcinoma prognosis (912). Although SPLUNC1-deficient mice do not develop spontaneous lung disease without a bacterial insult, they do exhibit more severe impaired intrapulmonary responses than wild-type controls when infected by respiratory pathogens such as Mycoplasma pneumoniae (13), Pseudomonas aeruginosa (14) and Klebsiella pneumoniae (15). In addition, loss of SPLUNC1 expression predisposes mice to develop otitis media (16). Although SPLUNC1 protein has been predicted to exert host defense and has been proposed as a valuable marker in non-small cell lung cancer (NSCLC) (17), the related immune cells regulated by SPLUNC1 remain elusive.

ALI triggers innate immune responses and is associated with the induction and promotion of immune-suppressive mechanisms, such as the accumulation of myeloid-derived suppressor cells (MDSCs). MDSCs have been described as a heterogenic population of immature myeloid cells that consist of myeloid progenitors and precursors of macrophages, granulocytes and dendritic cells (DCs) and that, mainly by multiple mechanisms, suppress T cell and NK cell functions (18,19). In healthy individuals, immature myeloid cells (IMCs) can be generated in bone marrow and differentiate into mature granulocytes, macrophages or DCs. In pathological conditions such as cancer, sepsis, various infectious diseases, trauma, certain autoimmune disorders or in bone marrow transplantation, a partial block in the differentiation of IMCs into mature myeloid cells results in an expansion of this population. In mice, MDSCs are identified as cells that co-express the myeloid lineage differentiation antigen Gr-1 and CD11b (20), which make up only a small proportion (2–4%) of spleen cells and are absent from the lymph nodes (18). Recently, several researchers have demonstrated that the accumulation of MDSCs plays a vital role in LPS-induced lung injury (21). However, the molecules that are involved in the generation and expansion of MDSCs during acute inflammation are not completely understood.

In the present study, we sought to investigate the effects of SPLUNC1 on lung injury using SPLUNC1-knockout (SPLUNC1−/−) mice, and to clarify the function of SPLUNC1- mediated recruitment of CD11b+Gr-1+ MDSCs in the spleen of LPS-induced ALI mice.

Materials and methods

Animals

To obtain SPLUNC1-knockout mice that allowed conditional and global disruption of the SPLUNC1 gene, we used the Cre/loxP and flp/FRT recombination systems to target exons 3 of SPLUNC1. A single loxp site was inserted before exon 3 and before exon 4, and an frt-flanked PGK neo cassette was inserted between exons 3 and 4 to serve as a positive selectable marker. The targeting construct was electroporated into embryonic stem (ES) cells and the germline SPLUNC1loxp-neo chimeras were obtained by homologous recombination. Then, the FRT-flanked PGK neo cassette was deleted by crossing female SPLUNC1loxp-neo mice with transgenic Flp male mice to obtain the homozygous mice for the floxed SPLUNC1 allele (SPLUNC1flox/flox). To generate SPLUNC1−/− mice, the SPLUNC1flox/flox mice were mated with EIIα-Cre mice (purchased from the Jackson Laboratory, Bar Harbor, ME, USA).

To produce animals for experiments, heterozygous mice were crossed to generate wild-type littermate controls. All mice used in the present study were male, 8–12 weeks old, and housed under specific pathogen-free conditions. The present study was performed in strict accordance with the recommendations of the Guide for the Care and Use of Laboratory Animals (National Institutes of Health). Experimental protocols were approved by the Animal Ethics Committee of Central South University.

Mouse model of LPS-induced ALI

Mice were randomly divided into four groups: WT control, KO control, WT-LPS, and KO-LPS. The LPS group was intraperitoneally injected with a single dose of 5 mg/kg body weight of LPS (Sigma, Carlsbad, CA, USA) which was dissolved in saline at a concentration of 1 mg/ml. Meanwhile, the mice in the control group were intraperitoneally injected with saline alone. The mice were euthanized at 24, 48, 72 and 96 h after LPS or saline administration.

Western blot analysis

The proteins were extracted from mouse lung tissues and then separated by 12% sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) and transferred to a nitrocellulose membrane. The membrane was blocked using 5% non-fat milk in Tris-buffered saline containing 0.1% Tween-20 and then incubated with primary antibodies: SPLUNC1 (R&D Systems, Minneapolis, MN, USA) and GAPDH (Santa Cruz Biotechnology, Inc., Santa Cruz, CA, USA) at 4°C overnight. After washing three times with TBST, the membranes were incubated with HRP-conjugated secondary antibody. Bands were detected with an electrochemiluminescence detection system.

Flow cytometric analysis

Single cell suspensions were prepared by homogenization through a sterile stainless steel mesh from the spleens. Erythrocytes were depleted using RBC lysing buffer (Sigma), and splenocytes were washed with phosphate-buffered saline (PBS). Then, the cells were stained with anti-CD11b and anti-Gr-1 antibodies (eBioscience, San Diego, CA, USA) according to the manufacturer's protocol. Fluorescence was measured using a MoFlo™ XDP High-Performance Cell Sorter and data were analyzed by Summit 5.2 software (both from Beckman Coulter, Inc., Brea, CA, USA).

RNA extraction and real-time PCR analysis

Reverse transcription reaction was performed using a Fermentas Revert Aid First Strand cDNA Synthesis kit (Fermentas, Burlington, ON, Canada), according to the manufacturer's instructions. qRT-PCR was performed using an iQ5 Multicolor Detection system (Bio-Rad, Hercules, CA, USA). The sequences of the primers were as follows: β-actin forward, 5′-TTTCCAGCCTTCCTTCTT-3′ and reverse, 5′-GGTCTTTACGGATGTCAACG-3′; IL-6 forward, 5′-CTGATGCTGGTGACAACCAC-3′ and reverse, 5′-CAGAATTGCCATTGCACAAC-3′; IL-8 forward, 5′-CGTCCCTGTGACACTCAAGA-3′ and reverse, 5′-GGAGCATCAGGATCCAAACA-3′; CCL-2 forward, 5′-ATGCAGTTAACGCCCCACTC-3′ and reverse, 5′-ACCCATTCCTTCTTGGGGTC-3′; CCL-3 forward, 5′-GCAACCAAGTCTTCTCAGCG-3′ and reverse, 5′-TTGGACCCAGGTCTCTTTGG-3′; CCL-5 forward, 5′-ACCATATGGCTCGGACACCA-3′ and reverse, 5′-GCGGTTCCTTCGAGTGACA-3′; CXCL1 forward, 5′-TGGCTGGGATTCACCTCAAG-3′ and reverse, 5′-CCGTTACTTGGGGACACCTT-3′; IL-1β forward, 5′-TGCCACCTTTTGACAGTGATG-3′ and reverse, 5′-AAGGTCCACGGGAAAGACAC-3′. The expression of mRNA was assessed by evaluated threshold cycle (CT) values. The CT values were normalized with the expression levels of β-actin and the relative amount of mRNA specific to each of the target genes was calculated using the 2−ΔΔCt method (2225). β-actin was used as an internal control.

Histology

Lungs of the differentially treated mice were fixed with 4% paraformaldehyde, and embedded in paraffin. Subsequently, the lung tissues were sliced into 4-µm sections, and then stained with hematoxylin and eosin to observe the inflammatory response and pathological changes using an Olympus BX51 microscope (Olympus, Tokyo, Japan). The degree of histologic lung injury was assessed based on the following criteria: neutrophil infiltration, alveolar congestion, interstitial edema, necrosis and atelectasis. The severity of injury was evaluated via a numerical rating system that ranges from 0 to 4 (score): no injury, 0; injury to 25% of the field, 1; injury to 50% of the field, 2; injury to 75% of the field, 3; and diffuse injury, 4. The mean score was then analyzed for comparison between groups.

Immunohistochemistry (IHC) and evaluation of staining

IHC was carried out using the peroxidase anti-peroxidase technique following a microwave antigen retrieval procedure. Anti-CD11b (MAB1124; 1:300), anti-Gr-1 (MAB1037; 1:300) (both from R&D Systems) was overlaid on the tissue sections and incubated overnight at 4°C. Secondary antibody incubation (Santa Cruz Biotechnology, Inc.) was performed at room temperature for 30 min. Color reaction was developed using 3,3′-diaminobenzidine tetrachloride (DAB) chromogen solution. All slides were counterstained with hematoxylin. Positive control slides were included in every experiment in addition to the internal positive controls. The specificity of the antibody was determined with matched IgG isotype antibody as a negative control.

Immunostaining was evaluated by two investigators in a blinded manner in an effort to provide a consensus on staining patterns by light microscopy (Olympus). CD11b or Gr-1 staining was assessed according to the methods described by Hara and Okayasu (26) with minor modifications. Each case was rated according to a score that added a scale of intensity of staining to the area of staining. At least 10 high-power fields were randomly chosen, and >1,000 cells were counted for each section. The intensity of staining was graded on the following scale: 0, no staining; 1+, mild staining; 2+, moderate staining; 3+, intense staining. The area of staining was evaluated as follows: 0, no staining of cells in any microscopic fields; 1+, <30% of tissue stained positive; 2+, between 30 and 60% stained positive; 3+, >60% stained positive. The minimum score when summed (extension + intensity) was, therefore, 0, and the maximum, 6. A combined staining score (extension + intensity) of ≤2 was considered to be a negative staining (low staining); a score between 3 and 4 was considered to be a moderate staining; whereas a score between 5 and 6 was considered to be a strong staining. An optimal cut-off level was identified as follows: a staining index score of 0–2 was used to define tumors with negative expression and 3–7 indicated positive expression of these two proteins. Agreement between the two evaluators was 95%, and all scoring discrepancies were resolved by discussion between the two evaluators.

Statistical analysis

The data obtained in the present study are expressed as the mean values ± standard deviation (SD) from at least three independent experiments using triplicate samples for the individual treatments. Statistical analyses were performed using two-way ANOVA or Fisher's exact test (Prism 6; GraphPad Software, San Diego, CA, USA). A P-value of <0.05 was considered to indicate a statistically significant difference.

Results

Generation of SPLUNC1-knockout mice

To identify the role of SPLUNC1 in vivo, we generated SPLUNC1-knockout mice. In this procedure, Cre/loxp and Flp/FRT recombination systems were used to target exon 3 of the mouse SPLUNC1 gene (Fig. 1A). The genomic DNA was extracted from the mouse tail and used as a template for PCR analysis with primer P1 and P2 to identify genotypes (Fig. 1B). SPLUNC1 protein expression in wild-type and SPLUNC1−/− mice was shown by western blot analysis (Fig. 1C). A specific SPLUNC1 band was detected in the SPLUNC1+/+ and SPLUNC1+/− lung, while no SPLUNC1 protein was detected in the lung of the SPLUNC1−/− mice. Then, we observed that knockout of SPLUNC1 did not cause embryonic death, and there was no obvious phenotype difference between the wild-type and SPLUNC1−/− mice. Similar results were reported by Gally et al (13) that SPLUNC1−/− mice were viable, fertile, and largely indistinguishable from WT littermates in general appearance, body weight, locomotion and overt behavior.

Knockout of SPLUNC1 affects the distribution of CD11b+Gr-1+ MDSCs in the spleen of LPS-induced ALI mice using flow cytometric analysis

MDSCs commonly express the markers CD11b and Gr-1 in mice; such immature myeloid cells are present in the bone marrow of healthy mice. However, they differentiate into mature myeloid cells and accumulate in the spleen under acute inflammation conditions. In the present study, we first measured the percentage of CD11b+Gr-1+ MDSCs in the bone marrow of the mice at different time points after LPS administration. The number of CD11b+Gr-1+ MDSCs in the WT group was reduced from 34.92±8.48 to 13.61±1.33% and in the KO group, from 37.65±8.06 to 12.81±5.92% (P<0.05; Fig. 2A) after LPS injection for 24 h, and then it rapidly recovered to the base level after injection for 48 h. Meanwhile, the number of these cells gradually increased at 72 and 96 h post LPS injection; it reached the highest level both in the WT and KO groups (70.98±2.84% in WT vs. 68.92±3.24% in KO mice at 96 h). Although the percentage of CD11b+ cells in KO mice was higher than that in WT mice at 24 h after LPS induction (P<0.05; Fig. 2B), no significant differences were observed between WT and KO groups at the different time-points of 0, 24, 48, 72 and 96 h after LPS injection (P>0.05; Fig. 2B). These results indicated that knockout of SPLUNC1 had no influence on the production of MDSCs in the bone marrow of the mice with ALI.

To investigate whether SPLUNC1 affects the recruitment of MDSCs from the bone marrow to the spleen in acute inflammatory response, we next analyzed the proportion of splenetic CD11b+Gr-1+ MDSCs in the WT and KO groups. There was significant accumulation of CD11b+Gr-1+ MDSCs after LPS injection, increasing from 3.26±0.41% of all splenocytes at baseline to a peak of 11.46±2.97% by 48 h in the WT group and from 3.44±0.46% of all splenocytes at baseline to a peak of 19.04±4.53% by 96 h in the KO group (Fig. 2C). The percentage of CD11b+ cells in the KO mice was higher than that in the WT mice at the time-points of 48 h (4.79±0.49% in WT mice vs. 8.67±2.38% in KO mice) and 96 h (3.44±0.74% in WT mice vs. 9.10±1.62% in KO mice) after LPS injection (P<0.05; Fig. 2D). Although the number of Gr-1+ cells in the WT mice was higher than that in the KO mice 96 h (15.18±2.87% in WT mice vs. 6.97±2.41% in KO mice) post LPS injection, the percentage of CD11b+Gr-1+ MDSCs in the KO mice was higher than that in the WT mice at the time point of 72 h (10.58±0.79% in WT mice vs. 16.36±2.42% in KO mice) and 96 h (10.95±1.11% in WT mice vs. 19.04±4.53% in KO mice) after LPS injection (P<0.05; Fig. 2D). These data revealed that the presence of SPLUNC1 suppresses the recruitment of CD11b+Gr-1+ MDSCs to the spleen in LPS-induced ALI.

Expression of CD11b/Gr-1 in cells in the mouse spleen as detected by IHC

Granulocyte differentiation antigen 1 (Gr-1) is highly expressed in neutrophils and Gr-1+ cells play a vital role in host defense, functioning as immunoregulators in the immune system. CD11b+ cells, specifically express M-CSF receptor (CD115) or IL-4R (CD124), having immunosuppressive activity. CD11b+ cells are distributed throughout the body and act as sentinels for the immune system. The frequency of CD11b+ myeloid cells varies in different organs, ranging from ~48% of brain myeloid (microglia) cells, 30% of bone marrow and 6% of total nucleated cells in the gut and spleen. In mice, MDSCs are identified as cells that co-express the myeloid lineage differentiation antigens Gr-1 and CD11b, which have been shown to accumulate quickly in mouse spleen and possess immunosuppressive effect in response to LPS administration. In the present study, we aimed to investigate whether SPLUNC1 affects the recruitment of MDSCs from the bone marrow to the spleen in acute inflammation; thus, it was important to distinguish between CD11b+Gr-1+ cells from CD11b+ and Gr-1+ cells.

In the present study, we examined the protein expression of Gr-1 and CD11b in the mouse spleen in response to LPS injection. In the WT and KO groups, few Gr-1+ cells were observed in the splenic tissues at 0 and 24 h after LPS injection, but a marked increase in Gr-1+ cells was noted by 48 h, particularly 72 and 96 h after LPS injection (Fig. 3A). Similarly, CD11b exhibited low expression at 0 and 24 h in cells of splenic tissues after LPS injection, while 48 h post injection, the number of cells expressing CD11b gradually increased. However, statistically significant differences between the WT and KO groups were not detected by IHC at the different time points after LPS injection (Fig. 3B).

Knockout of SPLUNC1 enhances LPS-induced lung injury

To investigate the role of SPLUNC1 in acute lung inflammation, WT and SPLUNC1−/− mice were treated with LPS as described in Materials and methods. The severity of lung injury was evaluated by histopathological analysis using hematoxylin and eosin (H&E) staining. In the WT and KO groups, the lungs of the mice injected with LPS showed neutrophil infiltration, thickened alveolar walls and lung congestion, while no such pathological changes were found in the control group (LPS-0 h). Meanwhile, when treated with LPS, the SPLUNC1−/− group appeared to have more severe pulmonary damage and infiltrated inflammatory cells compared with the WT mice after LPS treatment for 48 h (Fig. 4).

Knockout of SPLUNC1 promotes lung inflammation in response to LPS induction

Furthermore, we compared the differential expression of these cytokines in the WT and SPLUNC1−/− groups at different time points after LPS treatment. The mRNA expression levels of IL-6, CCL-2 and CCL-3 in the SPLUNC1−/− group were higher than these levels in the WT group 48 h post LPS injection. The level of CXCL-1 mRNA expression in the SPLUNC1−/− group was higher than the level in the WT group after LPS injection at 72 and 96 h (P<0.05; Fig. 5A). The SPLUNC1 expression levels were increased in the lung tissues of the WT mice after LPS injection (Fig. 5B). Taken together, these data demonstrated that SPLUNC1 deficiency upregulated ALI-related cytokines, which may promote early inflammation leading to more severe lung injury after LPS injection.

Discussion

The majority of recent studies on SPLUNC1 have attempted to explore its role in inflammation or infection, while the immune cells associated with SPLUNC1 regulation remain largely unknown. In the present study, SPLUNC1 protein was undetectable in SPLUNC1−/− mouse lung tissues, which confirmed gene knockout. Our daily observation found that there was no obvious phenotype difference between the wild-type and SPLUNC1−/− mice under normal physiological conditions, which was in accordance with a study reported by Gally et al (13).

SPLUNC1 plays a significant role in the host immune system with immunomodulatory (27), anti-biofilm (28) and/or chemotactic properties (29). Our in vitro studies have demonstrated that SPLUNC1 protein can bind to bacterial lipopolysaccharide (LPS), inhibit the growth of P. aeruginosa (10), regulate cell progression and apoptosis (12) and weaken the inflammatory response induced by EBV infection (11). To further explore its immune function in acute inflammation, we established an acute lung injury (ALI) mouse model by LPS intraperitoneal injection to WT and SPLUNC1-knockout mice, respectively. The symptoms of LPS-induced ALI mice closely resemble the observed pathology in humans (30); therefore, intraperitoneal injection of LPS has been widely used to study the pathogenesis of ALI.

ALI is a severe inflammatory disease that can lead to clinical syndromes including hypoxemia, pulmonary inflammation, alveolar-capillary barrier damage and multiple organ injury (2,31,32). LPS can be recognized by macrophages via their surface CD14/TLR4 receptor complexes. Activated macrophages and other immune cells then express multiple cytokines, which initiate, amplify and perpetuate the inflammatory response in ALI (33,34). As expected, the present study found that when treated with LPS, the SPLUNC1−/− group appeared to have more severe pulmonary damage compared with the WT group after LPS treatment. The characteristic of ALI is the release of chemokines, such as CCL-2, CCL-3 and CXCL-1, which recruit macrophage and neutrophil migration from the intravascular space across the endothelium and epithelium into the airspaces (35). In the present study, we found that the mRNA expression levels of IL-6, CCL-2, CCL-3 and CXCL-1 in the SPLUNC1−/− group were higher than these levels in the WT group at different time points after LPS injection and the period of increase of these cytokine in the KO group was longer than that in the WT group. Moreover, immunohistochemical results indicated that the expression of SPLUNC1 in the WT group also increased after LPS injection. These results demonstrated that SPLUNC1 deficiency upregulated ALI-related cytokines, which may promote early inflammation leading to more severe lung injury after LPS injection.

There are two types of innate immune molecular proteins that play a key role in the defense of gram-negative bacteria: lipopolysaccharide-binding protein (LBP) and bactericidal/permeability increasing protein (BPI). The two structures are similar, and all can be combined with LPS. LBP presents LPS to the surface of immune cells and stimulates them to produce an immune response. However, as an antagonist of LPS, BPI can block the combination of LPS and LBP (36,37). Our previous study found that SPLUNC1 has a similar folding structure to BPI and LBP, which can bind to bacterial LPS (10). Therefore, we concluded that the homology of the BPI domain of the SPLUNC1 molecule may be one of the important molecular basis of its protective function in LPS-induced lung injury in mice.

Various studies have suggested that Gr-1+/CD11b+ cells can play a vital role in the immune system and Gr-1+CD11b+ MDSCs have been shown to accumulate quickly in the mouse spleen and possess immunosuppressive effect in response to LPS administration (3841). In the present study, we found that the expression of those cells in mouse spleen was increased 48 h after LPS injection; the percentage of Gr-1+CD11b+ MDSCs in the spleen showed an increasing production trend post LPS injection both in the WT and KO groups. In the mouse bone marrow, the number of CD11b+Gr-1+ MDSCs declined 24 h post LPS injection, and then it recovered rapidly. Although there were no differences in mouse bone marrow between WT and KO groups after LPS injection, the percentage of splenic CD11b+Gr-1+ MDSCs in the KO mice was significant higher than that in the WT mice at the time-points of 72 and 96 h post LPS injection. Many factors can induce MDSC activation and expansion such as macrophage colony-stimulating factor (M-CSF), IL-6 (42), vascular endothelial growth factor (VEGF) (43), prostaglandins (44) and granulocyte/macrophage colony-stimulating factor (GM-CSF) (45). It has been reported that the molecules which are produced in lung injury can be linked to the bone marrow and show significant associations with the expansion and activation of MDSCs (46,47). These results indicate that SPLUNC1 can influence the recruitment of MDSCs by upregulating the secretion of cytokines in impaired lung tissue.

In summary, based on SPLUNC1-knockout mice we established the LPS-induced ALI mouse model to study the anti-inflammatory effect of SPLUNC1. Our results found that SPLUNC1 deficiency upregulated levels of ALI-related cytokines leading to more severe lung injury after LPS injection. Moreover, we first discovered that the presence of SPLUNC1 can suppress the recruitment of CD11b+Gr-1+ MDSCs to the spleen in LPS-induced ALI. Nevertheless, the mechanism of the CD11b+Gr-1+ MDSC recruitment needs to be further studied.

Acknowledgements

The present study was supported by the National Natural Sciences Foundation of China (nos. 81672685, 81402270, 81272975, 81672993 and 81402307), the Key Project of Hunan Provincial Natural Science Foundation (no. 12JJ2044), the Key Planned Science and Technology Project of Hunan Province (nos. 2012FJ2014 and 2011FJ3153); the 111 Project 111-2-12), the Natural Science Foundation of Hunan Province (2016JC2035), the Planned Project of Development and Reform Commission of Hunan Province (2012-1493-1), the Planned Project of Department of health of Hunan Province (B2011-030, B2012-029), the Open-End Fund for the Valuable and Precision Instruments of Central South University Doctoral innovation project of Hunan Province (CX2015B057).

Glossary

Abbreviations

Abbreviations:

SPLUNC1

short palate, lung and nasal epithelium clone 1

MDSCs

myeloid-derived suppressor cells

LPS

lipopoly-saccharide

ALI

acute lung injury

IMCs

immature myeloid cells

DCs

dendritic cells

IHC

immunohistochemistry

GADPH

glyceraldehyde-3-phosphate dehydrogenase

WT

wild-type

NSCLC

non-small cell lung cancer

IL-6

interleukin-6

CCL-2

chemokine (C-C motif) ligand-2

CCL-3

chemokine (C-C motif) ligand-3

CXCL-1

chemokine (C-X-C motif) ligand-1

References

1 

Rubenfeld GD, Caldwell E, Peabody E, Weaver J, Martin DP, Neff M, Stern EJ and Hudson LD: Incidence and outcomes of acute lung injury. N Engl J Med. 353:1685–1693. 2005. View Article : Google Scholar : PubMed/NCBI

2 

Abraham E: Neutrophils and acute lung injury. Crit Care Med. 31 Suppl 4:S195–S199. 2003. View Article : Google Scholar : PubMed/NCBI

3 

Bingle CD and Bingle L: Characterisation of the human plunc gene, a gene product with an upper airways and nasopharyngeal restricted expression pattern. Biochim Biophys Acta. 1493:363–367. 2000. View Article : Google Scholar : PubMed/NCBI

4 

Bingle CD and Craven CJ: Comparative analysis of the PLUNC (palate, lung and nasal epithelium clone) protein families. Biochem Soc Trans. 31:806–809. 2003. View Article : Google Scholar : PubMed/NCBI

5 

Zhang B, Nie X, Xiao B, Xiang J, Shen S, Gong J, Zhou M, Zhu S, Zhou J, Qian J, et al: Identification of tissue-specific genes in nasopharyngeal epithelial tissue and differentially expressed genes in nasopharyngeal carcinoma by suppression subtractive hybridization and cDNA microarray. Genes Chromosomes Cancer. 38:80–90. 2003. View Article : Google Scholar : PubMed/NCBI

6 

Kim CH, Kim K, Jik Kim H, Kim J Kook, Lee JG and Yoon JH: Expression and regulation of PLUNC in human nasal epithelium. Acta Otolaryngol. 126:1073–1078. 2006. View Article : Google Scholar : PubMed/NCBI

7 

Campos MA, Abreu AR, Nlend MC, Cobas MA, Conner GE and Whitney PL: Purification and characterization of PLUNC from human tracheobronchial secretions. Am J Respir Cell Mol Biol. 30:184–192. 2004. View Article : Google Scholar : PubMed/NCBI

8 

Sung YK, Moon C, Yoo JY, Moon C, Pearse D, Pevsner J and Ronnett GV: Plunc, a member of the secretory gland protein family, is up-regulated in nasal respiratory epithelium after olfactory bulbectomy. J Biol Chem. 277:12762–12769. 2002. View Article : Google Scholar : PubMed/NCBI

9 

Zhang W, Zeng Z, Wei F, Chen P, Schmitt DC, Fan S, Guo X, Liang F, Shi L, Liu Z, et al: SPLUNC1 is associated with nasopharyngeal carcinoma prognosis and plays an important role in all-trans-retinoic acid-induced growth inhibition and differentiation in nasopharyngeal cancer cells. FEBS J. 281:4815–4829. 2014. View Article : Google Scholar : PubMed/NCBI

10 

Zhou HD, Li XL, Li GY, Zhou M, Liu HY, Yang YX, Deng T, Ma J and Sheng SR: Effect of SPLUNC1 protein on the Pseudomonas aeruginosa and Epstein-Barr virus. Mol Cell Biochem. 309:191–197. 2008. View Article : Google Scholar : PubMed/NCBI

11 

Ou C, Sun Z, Zhang H, Xiong W, Ma J, Zhou M, Lu J, Zeng Z, Bo X, Chen P, et al: SPLUNC1 reduces the inflammatory response of nasopharyngeal carcinoma cells infected with the EB virus by inhibiting the TLR9/NF-κB pathway. Oncol Rep. 33:2779–2788. 2015. View Article : Google Scholar : PubMed/NCBI

12 

Chen P, Guo X, Zhou H, Zhang W, Zeng Z, Liao Q, Li X, Xiang B, Yang J, Ma J, et al: SPLUNC1 regulates cell progression and apoptosis through the miR-141-PTEN/p27 pathway, but is hindered by LMP1. PLoS One. 8:e569292013. View Article : Google Scholar : PubMed/NCBI

13 

Gally F, Di YP, Smith SK, Minor MN, Liu Y, Bratton DL, Frasch SC, Michels NM, Case SR and Chu HW: SPLUNC1 promotes lung innate defense against Mycoplasma pneumoniae infection in mice. Am J Pathol. 178:2159–2167. 2011. View Article : Google Scholar : PubMed/NCBI

14 

Lukinskiene L, Liu Y, Reynolds SD, Steele C, Stripp BR, Leikauf GD, Kolls JK and Di YP: Antimicrobial activity of PLUNC protects against Pseudomonas aeruginosa infection. J Immunol. 187:382–390. 2011. View Article : Google Scholar : PubMed/NCBI

15 

Liu Y, Bartlett JA, Di ME, Bomberger JM, Chan YR, Gakhar L, Mallampalli RK, McCray PB Jr and Di YP: SPLUNC1/BPIFA1 contributes to pulmonary host defense against Klebsiella pneumoniae respiratory infection. Am J Pathol. 182:1519–1531. 2013. View Article : Google Scholar : PubMed/NCBI

16 

Bartlett JA, Meyerholz DK, Wohlford-Lenane CL, Naumann PW, Salzman NH and McCray PB Jr: Increased susceptibility to otitis media in a Splunc1-deficient mouse model. Dis Model Mech. 8:501–508. 2015. View Article : Google Scholar : PubMed/NCBI

17 

Mitas M, Hoover L, Silvestri G, Reed C, Green M, Turrisi AT, Sherman C, Mikhitarian K, Cole DJ, Block MI, et al: Lunx is a superior molecular marker for detection of non-small cell lung cancer in peripheral blood [corrected]. J Mol Diagn. 5:237–242. 2003. View Article : Google Scholar : PubMed/NCBI

18 

Gabrilovich DI and Nagaraj S: Myeloid-derived suppressor cells as regulators of the immune system. Nat Rev Immunol. 9:162–174. 2009. View Article : Google Scholar : PubMed/NCBI

19 

Rabinovich GA, Gabrilovich D and Sotomayor EM: Immunosuppressive strategies that are mediated by tumor cells. Annu Rev Immunol. 25:267–296. 2007. View Article : Google Scholar : PubMed/NCBI

20 

Kusmartsev S, Nefedova Y, Yoder D and Gabrilovich DI: Antigen-specific inhibition of CD8+ T cell response by immature myeloid cells in cancer is mediated by reactive oxygen species. J Immunol. 172:989–999. 2004. View Article : Google Scholar : PubMed/NCBI

21 

Fu C, Jiang L, Xu X, Zhu F, Zhang S, Wu X, Liu Z, Yang X and Li S: STAT4 knockout protects LPS-induced lung injury by increasing of MDSC and promoting of macrophage differentiation. Respir Physiol Neurobiol. 223:16–22. 2016. View Article : Google Scholar : PubMed/NCBI

22 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2−ΔΔCT method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

23 

Zhou Y, Wang W, Zheng D, Peng S, Xiong W, Ma J, Zeng Z, Wu M, Zhou M, Xiang J, et al: Risk of nasopharyngeal carcinoma associated with polymorphic lactotransferrin haplotypes. Med Oncol. 29:1456–1462. 2012. View Article : Google Scholar : PubMed/NCBI

24 

Xiao S, Zhou Y, Yi W, Luo G, Jiang B, Tian Q, Li Y and Xue M: Fra-1 is downregulated in cervical cancer tissues and promotes cervical cancer cell apoptosis by p53 signaling pathway in vitro. Int J Oncol. 46:1677–1684. 2015. View Article : Google Scholar : PubMed/NCBI

25 

Zheng D, Liao S, Zhu G, Luo G, Xiao S, He J, Pei Z, Li G and Zhou Y: CD38 is a putative functional marker for side population cells in human nasopharyngeal carcinoma cell lines. Mol Carcinog. 55:300–311. 2016. View Article : Google Scholar : PubMed/NCBI

26 

Hara A and Okayasu I: Cyclooxygenase-2 and inducible nitric oxide synthase expression in human astrocytic gliomas: Correlation with angiogenesis and prognostic significance. Acta Neuropathol. 108:43–48. 2004. View Article : Google Scholar : PubMed/NCBI

27 

Thaikoottathil JV, Martin RJ, Di PY, Minor M, Case S, Zhang B, Zhang G, Huang H and Chu HW: SPLUNC1 deficiency enhances airway eosinophilic inflammation in mice. Am J Respir Cell Mol Biol. 47:253–260. 2012. View Article : Google Scholar : PubMed/NCBI

28 

Gakhar L, Bartlett JA, Penterman J, Mizrachi D, Singh PK, Mallampalli RK, Ramaswamy S and McCray PB Jr: PLUNC is a novel airway surfactant protein with anti-biofilm activity. PLoS One. 5:e90982010. View Article : Google Scholar : PubMed/NCBI

29 

Sayeed S, Nistico L, St Croix C and Di YP: Multifunctional role of human SPLUNC1 in Pseudomonas aeruginosa infection. Infect Immun. 81:285–291. 2013. View Article : Google Scholar : PubMed/NCBI

30 

Chen H, Bai C and Wang X: The value of the lipopolysaccharide-induced acute lung injury model in respiratory medicine. Expert Rev Respir Med. 4:773–783. 2010. View Article : Google Scholar : PubMed/NCBI

31 

Matuschak GM and Lechner AJ: Acute lung injury and the acute respiratory distress syndrome: Pathophysiology and treatment. Mo Med. 107:252–258. 2010.PubMed/NCBI

32 

Gaudry S, Ricard JD and Dreyfuss D: Acute respiratory distress syndrome. Rev Prat. 62:1197–1203. 2012.(In French). PubMed/NCBI

33 

Mei SH, McCarter SD, Deng Y, Parker CH, Liles WC and Stewart DJ: Prevention of LPS-induced acute lung injury in mice by mesenchymal stem cells overexpressing angiopoietin 1. PLoS Med. 4:e2692007. View Article : Google Scholar : PubMed/NCBI

34 

Zhang X, Sun CY, Zhang YB, Guo HZ, Feng XX, Peng SZ, Yuan J, Zheng RB, Chen WP, Su ZR, et al: Kegan Liyan oral liquid ameliorates lipopolysaccharide-induced acute lung injury through inhibition of TLR4-mediated NF-κB signaling pathway and MMP-9 expression. J Ethnopharmacol. 186:91–102. 2016. View Article : Google Scholar : PubMed/NCBI

35 

Bhatia M, Zemans RL and Jeyaseelan S: Role of chemokines in the pathogenesis of acute lung injury. Am J Respir Cell Mol Biol. 46:566–572. 2012. View Article : Google Scholar : PubMed/NCBI

36 

Kim JW, Gerwick L and Park CI: Molecular identification and expression analysis of two distinct BPI/LBPs (bactericidal permeability-increasing protein/LPS-binding protein) from rock bream, Oplegnathus fasciatus. Fish Shellfish Immunol. 33:75–84. 2012. View Article : Google Scholar : PubMed/NCBI

37 

Krasity BC, Troll JV, Weiss JP and McFall-Ngai MJ: LBP/BPI proteins and their relatives: Conservation over evolution and roles in mutualism. Biochem Soc Trans. 39:1039–1044. 2011. View Article : Google Scholar : PubMed/NCBI

38 

Vaknin I, Blinder L, Wang L, Gazit R, Shapira E, Genina O, Pines M, Pikarsky E and Baniyash M: A common pathway mediated through Toll-like receptors leads to T- and natural killer-cell immunosuppression. Blood. 111:1437–1447. 2008. View Article : Google Scholar : PubMed/NCBI

39 

Delano MJ, Scumpia PO, Weinstein JS, Coco D, Nagaraj S, Kelly-Scumpia KM, O'Malley KA, Wynn JL, Antonenko S, Al-Quran SZ, et al: MyD88-dependent expansion of an immature GR-1+CD11b+ population induces T cell suppression and Th2 polarization in sepsis. J Exp Med. 204:1463–1474. 2007. View Article : Google Scholar : PubMed/NCBI

40 

De Wilde V, van Rompaey N, Hill M, Lebrun JF, Lemaître P, Lhommé F, Kubjak C, Vokaer B, Oldenhove G, Charbonnier LM, et al: Endotoxin-induced myeloid-derived suppressor cells inhibit alloimmune responses via heme oxygenase-1. Am J Transplant. 9:2034–2047. 2009. View Article : Google Scholar : PubMed/NCBI

41 

McCubbrey AL, Barthel L, Mould KJ, Mohning MP, Redente EF and Janssen WJ: Selective and inducible targeting of CD11b+ mononuclear phagocytes in the murine lung with hCD68-rtTA transgenic systems. Am J Physiol Lung Cell Mol Physiol. 311:L87–L100. 2016. View Article : Google Scholar : PubMed/NCBI

42 

Bunt SK, Yang L, Sinha P, Clements VK, Leips J and Ostrand-Rosenberg S: Reduced inflammation in the tumor microenvironment delays the accumulation of myeloid-derived suppressor cells and limits tumor progression. Cancer Res. 67:10019–10026. 2007. View Article : Google Scholar : PubMed/NCBI

43 

Gabrilovich D, Ishida T, Oyama T, Ran S, Kravtsov V, Nadaf S and Carbone DP: Vascular endothelial growth factor inhibits the development of dendritic cells and dramatically affects the differentiation of multiple hematopoietic lineages in vivo. Blood. 92:4150–4166. 1998.PubMed/NCBI

44 

Serafini P, Carbley R, Noonan KA, Tan G, Bronte V and Borrello I: High-dose granulocyte-macrophage colony-stimulating factor-producing vaccines impair the immune response through the recruitment of myeloid suppressor cells. Cancer Res. 64:6337–6343. 2004. View Article : Google Scholar : PubMed/NCBI

45 

Bronte V, Chappell DB, Apolloni E, Cabrelle A, Wang M, Hwu P and Restifo NP: Unopposed production of granulocyte-macrophage colony-stimulating factor by tumors inhibits CD8+ T cell responses by dysregulating antigen-presenting cell maturation. J Immunol. 162:5728–5737. 1999.PubMed/NCBI

46 

Islam MN, Das SR, Emin MT, Wei M, Sun L, Westphalen K, Rowlands DJ, Quadri SK, Bhattacharya S and Bhattacharya J: Mitochondrial transfer from bone-marrow-derived stromal cells to pulmonary alveoli protects against acute lung injury. Nat Med. 18:759–765. 2012. View Article : Google Scholar : PubMed/NCBI

47 

Kotton DN, Ma BY, Cardoso WV, Sanderson EA, Summer RS, Williams MC and Fine A: Bone marrow-derived cells as progenitors of lung alveolar epithelium. Development. 128:5181–5188. 2001.PubMed/NCBI

Related Articles

Journal Cover

January-2018
Volume 39 Issue 1

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhang H, Li X, Liao S, Wang H, Chen P, Zhu G, Luo Y, Li G and Zhou Y: SPLUNC1 knockout enhances LPS-induced lung injury by increasing recruitment of CD11b+Gr-1+ cells to the spleen of mice. Oncol Rep 39: 358-366, 2018
APA
Zhang, H., Li, X., Liao, S., Wang, H., Chen, P., Zhu, G. ... Zhou, Y. (2018). SPLUNC1 knockout enhances LPS-induced lung injury by increasing recruitment of CD11b+Gr-1+ cells to the spleen of mice. Oncology Reports, 39, 358-366. https://doi.org/10.3892/or.2017.6063
MLA
Zhang, H., Li, X., Liao, S., Wang, H., Chen, P., Zhu, G., Luo, Y., Li, G., Zhou, Y."SPLUNC1 knockout enhances LPS-induced lung injury by increasing recruitment of CD11b+Gr-1+ cells to the spleen of mice". Oncology Reports 39.1 (2018): 358-366.
Chicago
Zhang, H., Li, X., Liao, S., Wang, H., Chen, P., Zhu, G., Luo, Y., Li, G., Zhou, Y."SPLUNC1 knockout enhances LPS-induced lung injury by increasing recruitment of CD11b+Gr-1+ cells to the spleen of mice". Oncology Reports 39, no. 1 (2018): 358-366. https://doi.org/10.3892/or.2017.6063