1
views
0
recommends
+1 Recommend
1 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Deletion of G protein-coupled receptor 116 enhances neutrophil function and aggravates lung injury in mice

      brief-report

      Read this article at

      ScienceOpenPublisherPMC
      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Acute lung injury (ALI) globally afflicts over 3 million individuals every year. It can eventually develop into acute respiratory distress syndrome (ARDS) with a high mortality of up to 40%. To date, ALI has been undertreated in terms of the feeble efficacy of clinical approaches and the lack of proven pharmacological targets. 1 G protein-coupled receptors (GPCRs), the promising targets of modern medicine, participate intensively in the regulation of human physiology and pathophysiology. Among them, Gpr116, which is expressed in alveolar epithelial cells and immune cells, has been reported to play a key role in maintaining alveolar homeostasis and inhibiting inflammation. 2 However, its role in the regulation of neutrophil function and ALI remains to be elucidated. In this study, we demonstrated the inhibitory role of Gpr116 on neutrophil function, which contributes to nonspecific lung injury in mice induced by lipopolysaccharide (LPS). In brief, LPS was intratracheally injected at a dose of 10 mg/kg in mice to induce ALI in vivo. Neutrophils isolated from the bone marrow of control and myeloid cell-specific Gpr116 knockout mice were stimulated with phorbol 12-myristate 13-acetate (PMA) (100 ng/mL) in vitro. More details are available in the Supplementary materials. In clinical data, Gpr116 mRNA expression in blood samples was lower in patients with ARDS than in non-ARDS patients without statistical significance (Fig. 1A). Furthermore, we detected whether Gpr116 expression was altered in ALI mice. After administering LPS to C57BL/6J mice, we measured Gpr116 expression in lung tissue and BALF cells. Compared with that in control mice, the mRNA expression of Gpr116 in lung tissues and BALF cells of ALI mice decreased (Fig. 1B, C), along with similar changes in the protein expression of Gpr116 in the mouse lungs (Fig. S1). Besides, in the in vitro experiment, the mRNA expression of Gpr116 in bone marrow-derived neutrophils also decreased after PMA stimulation (Fig. S2). More significantly, Gpr116 expression in the representative immunofluorescence images was apparently downregulated and negatively correlated with the MPO-positive cells in the lungs of ALI mice compared to that of control mice (Fig. S3). These data indicate that Gpr116 expression is downregulated in lung injury, suggesting that it may play a role in the progression of ALI. Figure 1 The pathological effect of Gpr116 deletion in ALI mice and primary neutrophils. (A) Gpr116 mRNA levels in human blood samples based on the available GEO dataset (GSE 32707; t-tests). (B) Gpr116 mRNA levels in mouse lungs (n = 5; Student's t-test). (C) Gpr116 mRNA levels in BALF cells of mice (n = 5; Student's t-test). (D) Representative histopathological images (scale bars: 200 μm) and injury scores of mouse lungs (n = 5; one-way analysis of variance). (E) Total protein concentrations in mouse BALF (n = 3 for control and DNase I mice; n = 5 for LPS and DNase I + LPS mice; one-way analysis of variance). (F) Lung wet-to-dry (W/D) weight ratios (n = 5; one-way analysis of variance). (G) Survival curves of mice challenged with LPS (40 mg/kg, i.t.) (n = 5 for control mice; n = 8 for LPS mice; log-rank test). (H) Plasma concentrations of TNF-α and IL-6 (n = 5; one-way analysis of variance). (I) BALF concentrations of TNF-α and IL-6 (n = 5; one-way analysis of variance). (J) BALF concentrations of neutrophils quantified by flow cytometry (n = 5; one-way analysis of variance). (K) Plasma levels of cf-DNA (n = 5; one-way analysis of variance). (L) BALF levels of cf-DNA (n = 5; one-way analysis of variance). (M) Representative immunofluorescence images of NETs formation in mouse lungs (DAPI: blue; Cit-H3: red; MPO: green; DAPI + Cit-H3 + MPO + NETs are indicated by white arrowheads; scale bars: 50 μm). (N) TNF-α and IL-6 concentrations in the supernatants of bone marrow neutrophils from mice (n = 5; one-way analysis of variance). (O) cf-DNA concentrations in the supernatants of bone marrow neutrophils from mice (n = 5; one-way analysis of variance). (P) Representative immunofluorescence images of NETs formation in bone marrow-derived neutrophils (DAPI: blue; Cit-H3: red; MPO: green; the white arrow indicates DAPI  +  Cit-H3 + MPO  +  NETs; scale bars: 50 μm). Data are expressed as mean ± SEM of three repeated trials; ∗P < 0.05, ∗∗P < 0.01, #P < 0.05, ##P < 0.01. Figure 1 Accumulating evidence has shown that macrophages and neutrophils, the main effector cells of innate immunity, are recruited into the blood and migrate to alveolar capillaries along the gradient of chemoattractant. Both of them can release a variety of bactericidal mediators to induce diffuse damage of the blood-gas barrier and increased respiratory load, resulting in fatal hypoxemia. 3 Therefore, we bred myeloid cell-specific Gpr116 knockout mice using conditional CRE/LoxP deletion approaches (Fig. S4, S5) to further verify the role of Gpr116 in immune regulation in ALI. As shown by H&E staining, compared to flox mice, Gpr116 cKO mice were more sensitive to LPS challenge and had a higher lung injury score (Fig. 1D). In addition, the pathological damage of ALI also involves rupture of alveolar capillary membranes and leakage of protein-rich fluid in the alveoli, leading to pulmonary edema, which could be verified by the protein levels in BALF and lung W/D ratios. Here, we observed a dramatic increase of protein level in the BALF of Gpr116 cKO mice compared to flox mice (Fig. 1E). Likewise, the W/D ratio of lung tissues was also greater in cKO mice than that in flox mice (Fig. 1F). Furthermore, deletion of Gpr116 reduced mouse survival rate after LPS stimulation (40 mg/kg, i.t.) (Fig. 1G). In addition, we investigated the effect of Gpr116 on LPS-induced inflammation. Compared with flox mice, BALF and plasma concentrations of TNF-α and IL-6 in cKO mice increased after LPS injection, indicating that Gpr116 may inhibit pulmonary and systemic inflammation, although not all these discrepancies were statistically significant (Fig. 1I, H). These results are consistent with the reported role of Gpr116 in immune regulation, for example, Gpr116 suppresses inflammatory responses of alveolar macrophages via NF-κB signaling. 2 In fact, lung is thought to be main reservoir of neutrophils, and neutrophil functions involving recruitment, reactive oxygen species (ROS) production, and NETs release are closely associated with the pathogenesis of ALI. Nevertheless, the role of Gpr116 in neutrophil function remained to be illuminated. Here, we observed a dramatic increase in neutrophil lung infiltration in Gpr116 cKO mice compared to flox mice (Fig. 1J; Fig. S7). It has been reported that NETs, the new mechanisms of neutrophil immunity, are large, extracellular, web-like structures composed of cytosolic and granular proteins that are assembled on a scaffold of decondensed chromatin. Excessive NETs formation, not only a pathological result but also a pathogenic factor, plays an important role in ALI. 4 , 5 Therefore, we speculated that the protective effect of Gpr116 on ALI may be achieved by regulating the release of NETs. Subsequently, PicoGreen assays, Western blotting and immunofluorescence staining were performed to detect the marker levels of NETs involving cf-DNA, citrullinated histone H3 (Cit-H3), and MPO. As expected, we found that Gpr116 deletion dramatically increased the release of NETs in ALI mice (Fig. 1K–M; Fig. S8–9). ROS production in mouse lungs was also detected to further explore the regulatory effect of Gpr116 on neutrophil function. As Figure S6 shows, Gpr116 deficiency dramatically increased the production of ROS in ALI mouse lungs. DNase I is a specific inhibitor of NETs and can reduce NETs-associated citrullinated histones and minimize immune cell recruitment. To further confirm the inhibitory effects of Gpr116 on NETs release, we eliminated the formation of NETs by DNase I pretreatment before LPS stimulation. It was observed that specific inhibition of NETs reversed the aggravation of lung injury induced by Gpr116 deletion, as indicated by the reduced lung injury score (Fig. 1D), as well as the decreased protein levels in BALF and lung W/D ratios (Fig. 1E, F). Furthermore, the ELISA assays illustrated that the exacerbation of pulmonary and systemic inflammatory responses following Gpr116 deletion in ALI mice was abrogated by DNase I preintervention, as evidenced by the decreased TNF-α and IL-6 levels in BALF (Fig. 1I) and plasma (Fig. 1H), respectively. Taken together, these data suggest that Gpr116 may protect mice against lung injury by inhibiting neutrophil recruitment, NETs release, ROS production and inflammatory response. Rather, we cannot exclude the pathogenic role of macrophages during the progression of ALI. Therefore, we carried out magnetic isolation (Fig. S10) of neutrophils from bone marrow of control and conditional CRE/LoxP deletion of Gpr116 mice to further investigate the molecular mechanism of protective role of Gpr116. Consistent with the in vivo data, the NETs release of neutrophils from Gpr116 cKO mice increased notably after PMA challenge (Fig. 1O, P). Besides, the elevation of TNF-α and IL-6 levels in neutrophil supernatant induced by Gpr116 deletion was remarkable (Fig. 1N). These results elucidate the suppressive role of Gpr116 in neutrophil function, which potentially contributes to diffuse damage of blood-air barrier in an ALI mouse model. In conclusion, our data delineate that GPR116 can inhibit neutrophilic lung infiltration, NETs release, ROS production and inflammatory response to alleviate lung injury and mortality in ALI mice, which extends the therapeutic potential of GPR116. Further studies are needed to clarify the underlying mechanism of the regulation of GPR116 in neutrophil function to provide compelling evidence that GPR116 is a promising therapeutic target in alleviating ALI. Author contributions T Wang and JJ Bian designed the research; T Wang, Y Wang, and Q Xiang performed the research and analyzed the data; SW Lin, PP Jin, J Wang, and N Li offered technical support; T Wang, JF Wang and JJ Bian drafted and revised the manuscript. Conflict of interests The authors declare no conflict of interests. Funding This work was supported by the 10.13039/501100001809 National Natural Science Foundation of China (No. 81871579) and the Military Medical Science and Technology Youth Cultivate Program (China) (No. 19QNP018). Ethics declaration All experimental procedures were approved by the Laboratory Animal Ethics Committee of the Naval Medical University and were performed according to the relevant guidelines. Availability of data and materials The datasets used in this study are available from the corresponding author on reasonable request.

          Related collections

          Most cited references5

          • Record: found
          • Abstract: found
          • Article: found

          Acute Respiratory Distress Syndrome

            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Maladaptive role of neutrophil extracellular traps in pathogen-induced lung injury

            Neutrophils dominate the early immune response in pathogen-induced acute lung injury, but efforts to harness their responses have not led to therapeutic advancements. Neutrophil extracellular traps (NETs) have been proposed as an innate defense mechanism responsible for pathogen clearance, but there are concerns that NETs may induce collateral damage to host tissues. Here, we detected NETs in abundance in mouse models of severe bacterial pneumonia/acute lung injury and in human subjects with acute respiratory distress syndrome (ARDS) from pneumonia or sepsis. Decreasing NETs reduced lung injury and improved survival after DNase I treatment or with partial protein arginine deiminase 4 deficiency ( PAD4 +/– ). Complete PAD4 deficiency ( PAD4 –/– ) reduced NETs and lung injury but was counterbalanced by increased bacterial load and inflammation. Importantly, we discovered that the lipoxin pathway could be a potent modulator of NET formation, and that mice deficient in the lipoxin receptor ( Fpr2 –/– ) produced excess NETs leading to increased lung injury and mortality. Lastly, we observed in humans that increased plasma NETs were associated with ARDS severity and mortality, and lower plasma DNase I levels were associated with the development of sepsis-induced ARDS. We conclude that a critical balance of NETs is necessary to prevent lung injury and to maintain microbial control, which has important therapeutic implications. Neutrophil extracellular traps (NETs) are released after bacterial pneumonia in mice and in humans with acute respiratory distress syndrome, and contribute to lung barrier disruption.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Respiratory drive in the acute respiratory distress syndrome: pathophysiology, monitoring, and therapeutic interventions

              Neural respiratory drive, i.e., the activity of respiratory centres controlling breathing, is an overlooked physiologic variable which affects the pathophysiology and the clinical outcome of acute respiratory distress syndrome (ARDS). Spontaneous breathing may offer multiple physiologic benefits in these patients, including decreased need for sedation, preserved diaphragm activity and improved cardiovascular function. However, excessive effort to breathe due to high respiratory drive may lead to patient self-inflicted lung injury (P-SILI), even in the absence of mechanical ventilation. In the present review, we focus on the physiological and clinical implications of control of respiratory drive in ARDS patients. We summarize the main determinants of neural respiratory drive and the mechanisms involved in its potentiation, in health and ARDS. We also describe potential and pitfalls of the available bedside methods for drive assessment and explore classical and more “futuristic” interventions to control drive in ARDS patients.
                Bookmark

                Author and article information

                Contributors
                Journal
                Genes Dis
                Genes Dis
                Genes & Diseases
                Chongqing Medical University
                2352-4820
                2352-3042
                24 September 2022
                July 2023
                24 September 2022
                : 10
                : 4
                : 1214-1216
                Affiliations
                [1]Department of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai 200433, China
                Author notes
                []Corresponding author. Fax: +86 21 31161841. jinjunbicu@ 123456163.com
                [1]

                These authors contributed equally to this work.

                Article
                S2352-3042(22)00238-0
                10.1016/j.gendis.2022.08.017
                10311051
                956aca01-5417-4938-a5f5-5dea3de54dac
                © 2022 The Authors. Publishing services by Elsevier B.V. on behalf of KeAi Communications Co., Ltd.

                This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

                History
                : 27 March 2022
                : 30 July 2022
                : 15 August 2022
                Categories
                Rapid Communication

                Comments

                Comment on this article