INTRODUCTION
Chronic obstructive pulmonary disease (COPD) is a chronic systemic inflammatory disease characterized by airflow obstruction, pulmonary function deterioration, cough with sputum, and shortness of breath, which is progressive and not completely reversible (
1,
2). It is a major cause of morbidity and disability worldwide and is considered the third leading cause of death globally (
3,
4). Periodontitis is a chronic infectious disease caused by dental plaque biofilm and acts as a potential risk factor for a variety of systemic diseases, such as cardiovascular disease, diabetes, hypertension, various cancers, and chronic obstructive pulmonary disease (
5–9). Periodontitis is a microbial biofilm-associated infectious disease.
Porphyromonas gingivalis (P. gingivalis) is related to the severity of periodontal disease and has been identified as one of the critical virulence factors of periodontitis (
10). Even at low abundance,
P. gingivalis can induce chronic periodontitis through interference with host immune responses and causing dysbiosis of the oral commensal bacterial community (
10–12), and recently, it has been identified as the keystone bacteria in the dysbiosis of oral microbiota and periodontitis occurrence and development (
10–12).
Oral health status seriously impacts respiratory diseases via several aspects (
13–15): (i) Changes in oral health status leads to oral immune imbalance, after which local oral immune cells migrate to the lung, causing an immune imbalance of lung tissue and, thus, affect lung diseases; (ii) Changes in oral health status can also promote the progression of lung disease by affecting systemic inflammation. (iii) Oral health status is closely related to changes of oral microbiota, and then oral microorganisms affect the lung flora through inhalation and colonization on lung tissue, leading to lung immune imbalance and inflammatory response, and affecting the progression of lung diseases. At present, the lung microbiota changes and immune imbalance caused by ectopic colonization of oral microorganisms in the lungs is one of the main ways that oral health status affects respiratory diseases. Studies have shown that
P. gingivalis in the mouth of periodontitis patients or experimental mice can transfer and colonize on the lungs, causing lung inflammation and injury (
16–18). Periodontal pathogens such as
P. gingivalis,
Fusobacterium. nucleatum,
Prevotella oralis, and
Aggregatibacter actinomycetemcomitans have been identified in the lung aspiration of pneumonia patients (
2). Bronchial microbial studies of asthma indicated the presence of four specific bacterial groups, two of which,
Fusobacterium and
Porphyromonas, are known periodontal pathogenic bacteria (
19). Similarly, higher diversity and rates of microbial colonization were found in the lungs of patients with COPD, and microorganisms from the mouth and upper respiratory tract were detected (
2,
20). Previous studies have indicated that the oral periodontitis-associated bacteria
P. gingivalis acted as one of the main causes of the life-threatening infectious disease aspiration pneumonia and that
P. gingivalis gingipains could lead to the upregulation of inflammatory factors, including TNF, IL-6, IL 17, and C-reactive protein (
21). In short, it indicated that there was a microbial link between periodontitis and COPD state.
Dozens of studies have confirmed that there is, indeed, a positive correlation between periodontitis and COPD (
22,
23). People with severe periodontitis have a much higher relative risk of COPD compared with periodontal healthy counterparts (
24). The frequency of lung function attenuation and exacerbation in COPD patients with periodontitis is more serious than in COPD patients with good periodontal health (
25). Through basic periodontal treatment and oral health maintenance, the frequency of lung function attenuation and exacerbation in COPD patients can be well controlled (
26,
27). However, the specific mechanism by which periodontitis affects COPD progression still requires further exploration.
γδ T cells are T cells that perform innate immune functions, which could secret IL 17 and initiate inflammatory responses (
28). Studies have shown that oral microorganisms can promote the proliferation of
in situ γδ T cells in the lungs and secretion of IL 17 to mediate lung inflammation and disease status (
29,
30). However, the effect of γδ T cells on COPD is controversial and needs further study. Under the stimulation of IL 4/IL 10/IL 13, macrophages could become M2-polarized, after which cytokines such as MMP9 and MMP12 are produced by M2-polarized macrophages to promote the process of parenchymal injury (
31,
32). The occurrence of COPD is related to the enhancement of immune response mediated by macrophage polarization (
33,
34). The correlation between M2 polarization and COPD is controversial, and the mechanism of M2 polarization in periodontitis-promoted COPD remains unclear and needs further study. Through chemokines, cytokines, and cell surface receptors, γδ T cells and macrophages may undergo some inter-regulation (
35–37). IL 17 and IFN γ produced by γδ T cells can play an inflammatory role; meanwhile, IL 17 can also promote the M2 polarization of lung macrophages (
38,
39).
Based on the above background, we hypothesized that in the periodontitis state, the major periodontal pathogenic bacteria, as represented by P. gingivalis, may transfer and colonize on the lung tissue, causing microbiota changes in the lung, further leading to pulmonary immune imbalance and promoting the accumulation and proliferation of γδ T cells in the lung. On the one hand, γδ T cells produce IL 17 and IFN γ to directly promote the COPD process; on the other hand, the production of IL 17 by γδ T cells further promotes the M2 polarization of alveolar macrophages and contributes to COPD progression. Therefore, this study intends to determine whether periodontitis promotes COPD progression by regulating γδ T cells and M2 macrophages through a series of experiments, including in vivo animal models and ex vivo bacterial cell co-culture, and clinical sample detection, in order to provide new directions for clinical COPD patient management and disease control from the perspective of oral health.
DISCUSSION
Periodontitis is a chronic inflammatory disease of the periodontal tissue caused by plaque biofilm, which acts as a potential risk factor for a variety of systemic diseases, including cardiovascular disease, diabetes, hypertension, various cancers, COPD, etc. (
5,
42,
43). As is commonly known, globally, chronic respiratory diseases act as the leading cause of death and morbidity, among which COPD causes the most deaths, and is considered the third leading cause of death worldwide (
44). COPD is a devastating lung disease characterized by an incomplete and reversible airflow restriction that is usually progressive and associated with the lungs' abnormal inflammatory response to harmful particles or gases (
45). Many previously published studies reported the positive association between periodontitis and COPD (
22,
23), but the potential immune mechanisms were unclear.
In the current study, we found that periodontitis could aggravate COPD progression (
Fig. 1), which was consistent with previous studies (
1,
46). Further microbe analysis indicated that microbiota composition changed under disease state, and that periodontitis caused an increase of lung inflammation associated bacteria, such as
Proteobacteria and
Ralstonia. To some degree, this phenomenon indicated that periodontitis could promote lung inflammation development. Like previous studies (
2,
19,
20), we also observed the microorganism communication between lung tissue and the oral cavity. Many oral bacteria were detected in the lung tissues, including
Lactobacillus, Prevotella,
Bactriodes, and
P. gingivalis. Periodontitis also caused the increase of two main bacterial taxa,
Ralstonia and
Pelomonas, in the COPD lungs. Moreover, some oral periodontal-associated pathogenic bacteria, including
Prevotella,
Bactriodes, and
P. gingivalis were also detected with increased relative abundance in the COPD with periodontitis lung tissue compared to those in the COPD group. Therefore, it could be concluded that periodontitis affected the microbiota homeostasis of the lung.
γδ T cells perform innate immune functions and act as an important subset of T cells, which play an important role in inflammatory immunity via the secretion of cytokines, including IFN γ and IL 17 (
28). IL 17 is a pivotal cytokine that regulates lung immunity and inflammation and has shown the capacity to mediate COPD progression (
47,
48). IFN γ is another crucial cytokine that is associated with COPD. Numerous studies demonstrated that the IFN γ levels as well as IFN γ-producing cells were increased in COPD patients and experimental mouse models, while effective COPD treatment resulted in decreased IFN γ levels (
49–51). Moreover, a recent study indicated that Th17/Th1 cells capable of secreting both IL 17 and IFN γ were more pathogenic in COPD (
52).
In lung tissue, γδ T cells constitute a major tissue-resident T cell component and play important roles in maintaining the homeostasis of lung tissue (
53,
54). The effects of γδ T cells have two sides in lung tissue. Some studies have indicated that γδ T cells helped to activate host defense against bacterial, viral, and fungal infections and participated in maintaining lung tissue homeostasis (
53,
54). Under inflammation state of lung tissue, γδ T cells displayed a tissue reparative effect through the production of IL 17 and were found to be associated with inflammation control (
54,
55). Additionally, γδ T cells in lung tissue also showed protective effects towards cystic fibrosis and lung cancers (
53,
56,
57). However, on the contrary, numerous other studies have indicated that γδ T cells might act with a crucial pathogenic effect in lung diseases. γδ T cells and their IL 17 expression are required for pulmonary inflammation and injury caused by ozone exposure (
30). IL 17A-producing γδ T and Th17 lymphocytes mediated lung inflammation and acute lung injury in experimental silicosis (
58). Jin et al. (
29) implicated that commensal-bacteria activated γδ T cells in lung tissue and promoted lung adenocarcinoma through IL 17 production. Data in Vigeland’s study elucidate the critical role of IL 17A
+ γδ T cells in promoting chronic inflammation and fibrosis of the lung tissue (
39).
Similarly, there has been some controversy about the role of γδ T cells in COPD progression. Pons et al. (
59) reported blunted γδ T-lymphocyte response in the COPD disease state. Similarly, Urboniene et al. (
60) investigated γδ T cells in induced sputum (IS) and bronchoalveolar lavage (BAL) and found reduced amount of γδ T cells in IS and BALF from COPD patients compared those from asthmatic or healthy subjects, which indicated that γδ T cells might have a protective effect in preventing COPD. However, inconsistent with this, Majo et al. observed in clinical lung tissue samples that γδ T cells significantly increased in the lungs of smoker and emphysema patients (
61). Smoking causes or exacerbates COPD by promoting γδ T proliferation and IL 17 production (
48). Serum amyloid A promotes lung neutrophil accumulation and COPD by increasing γδ T cells and the associated expression of IL 17 (
62). In this study, we found that γδ T cells played an important role in the cigarette smoke exposure-induced COPD disease model, suggesting that smoking led to the expansion of γδ T cells in lung tissue and contributed to the occurrence of COPD (
Fig. 3 and 5), consistent with Majo’s and Bozinovski’s results (
48,
61). More importantly, we further found that γδ T cells were important targets for mediating the promotive effect of periodontitis on COPD. In the presence of periodontitis, the severity of COPD disease was much greater, the proportion of γδ T/IL17
+ γδ T/IFN γ
+ γδ T cells in COPD lung tissue was significantly up-regulated, and the contents of IL 17 and IFN γ were also significantly increased (
Fig. 3; Fig. S1 and S2). However, when γδ T cells were inhibited, the proportion of γδ T/IL 17
+ γδ T/IFN γ
+ γδ T cells in lung tissue was significantly reduced, the contents of IL 17 and IFN γ were decreased, the severity of COPD disease was significantly decreased, and the promotive effect of periodontitis on COPD was also significantly affected (
Fig. 5; Fig. S8). Therefore, these results suggested that periodontitis contributed to COPD progression by promoting γδ T cells activation in COPD lung tissue.
Macrophages are innate immune cells and play important roles in homeostasis and host defense. The development of COPD is associated with enhanced immune response mediated by macrophage polarization (
33,
34,
63,
64). A majority of alveolar macrophages are normally non-polarized in healthy lung tissue, but in lung tissue of COPD, there is a significant increase of macrophage polarization and co-expression of M1 and M2 macrophages (
65,
66). Moreover, both M1-polarized and M2-polarized macrophages in the lung tissue increased significantly with the progression of smoke and COPD severity (
65). However, the role of distinct macrophage phenotypes (M1 versus M2) in COPD is unclear and which macrophage phenotype (M1 versus M2) is more predominant in COPD progression is still controversial. Through literature review, Lee et al. (
64) concluded that M1-associated marker (iNOS) and cytokines (IL-1β, IL-6, IL-8, TNF-α) were increased in COPD patients, indicating the pathogenic effect of M1 polarization in COPD. LncRNA MIR155HG promoted COPD by upregulating M1 polarization and downregulating M2 polarization of lung macrophages (
67). Similarly, Sun’s data indicated ergosterol showed therapeutic effect toward COPD by decreasing M1 polarization and increasing M2 polarization (
68). While, dozens of studies have pointed out that M2 polarization of macrophages was the essential pathogenesis of COPD. Eapen et al. (
69) observed that in COPD patients, luminal macrophages showed a dominant polarization of M2 phenotype and BALF showed increased cytokines of the M2 profile, including CCL22, IL-4, IL-13, and IL-10. Liu et al. (
70) emphasized the important role of M2 polarization of macrophage and suggested that through suppressing M2 macrophage polarization, effective components combination (ECC) could improve airway remodeling of COPD. In the current study, we found that periodontitis promoted macrophage polarization in COPD lung tissue, including M1 polarization and M2 polarization, and the effect of periodontitis on M2 polarization in COPD lung tissue was significantly enhanced with the disease duration, which is consistent with the study of Feng et al. (
71), while, M1 polarization did not change significantly with the disease duration. Therefore, we assumed that in the later stage of the disease, M2 polarization played an important role in mediating the promotive effect of periodontitis on COPD.
Generally, IL 4, IL10, and IL 13 are considered the more common markers for inducing M2 polarization; however, existing studies have shown that in the lung tissue, γδ T cells could regulate M2 polarization of macrophages by producing IL17 and that M2 polarization could be significantly inhibited if γδ T cells was deficient or IL17 was blocked. In lung tissue, IL 17 expressed by activated γδ T cells could induce macrophage proliferation and M2 polarization, thus further affect disease progression (
30,
38,
39). This procedure had been demonstrated in both pulmonary inflammation and lung fibrosis (
38,
39) as well as in OSCC with periodontitis models (
35). Therefore, does such an immune regulatory mechanism also exist in the COPD disease model and can periodontitis further promote COPD progression by regulating this mechanism? In the current study, we found that M2 polarization was influenced by the activation of γδ T cells in lung tissues in COPD with periodontitis. The increase of γδ T and IL17
+ γδ T cells could further induce the increase of M2 polarization (
Fig. 4 and 5). Similarly, under the condition of γδ T cells inhibition treatment, γδ T and IL17
+ γδ T cells were significantly reduced, and M2 polarization was also significantly decreased (
Fig. 4 and 5). While, under γδ-TCR monoclonal antibody treatment, M1 polarization of macrophages in the lungs was not significantly affected, regardless of the presence or absence of periodontitis (Fig. S7). These results again demonstrated that the activation of γδ T cells in the lung tissue could partly induce the M2 polarization of macrophages and promote disease progression in the periodontitis-promoted COPD model. Importantly, in clinical BALF samples, we obtained similar results. In COPD BALF samples containing the periodontal pathogenic bacteria
P. gingivalis, the levels of IL 17 and IFN γ and the gene expressions of
IL 17,
IFN γ, and M2 polarization associated genes, including
ARG1,
CD206,
MMP9,
MMP12,
TGF β,
IL 4, and
IL10, were significantly upregulated (
Fig. 6), indicating the important roles of IL 17 and IFN γ and M2 macrophages in periodontitis-aggravated COPD in the clinical samples. While, in the disease models of current research, the regulatory mechanism of M1 polarization may have a more complex regulatory network, which needs to be further explored in the future. In the current disease model, periodontitis promoted the activation of γδ T cells in the lung. On the one hand, IFN γ and IL17 produced by activated γδ T cells directly promoted the progression of COPD. On the other hand, IL17 produced by activated γδ T cells further promoted M2 polarization, thereby aggravating the progression of COPD. However, in this disease model, M1 polarization might be regulated by a variety of inflammatory and immune factors, not just IFN γ. And in our future studies, we will further explore the regulatory network of M1 polarization in this disease model. Moreover, a novel concept mentioned that some novel markers (
CD38,
Gpr18, and
Fpr2 for M1 polarization,
Egr2 and
c-Myc for M2 polarization) (
72) were recommended to be contained in the macrophage polarization-associated studies, and in the future’s experimental study, we will further contain these markers.
Through the
in vivo,
ex vivo, and clinical samples analysis, we could conclude that in COPD with periodontitis, the microbiota homeostasis of lung tissue changed, and at the same time, an immune imbalance of lung tissue occurred. Periodontitis promoted the activation of γδ T cells, leading to γδ T cells, IL17
+ γδ T cells, and IFN γ
+ γδ T cells expansion in the COPD lung tissue. On the one hand, IL 17 and IFN γ directly promoted the progression of COPD, and, simultaneously, IL 17 further induced M2 polarization of macrophages, thereby contributing to COPD progression (
Fig. 7).
However, there are some limitations in this research. The first one is the short duration of the later-stage model. In this study, the later-stage model (4-week model) only extended the cigarette smoke exposure time by 2 weeks compared to the earlier-stage model (2-week model), which might be related to the fact that in this research, the decrease of the lung functions and the increase of some immune cells (γδ T cells and M2 macrophages) were modest. If the cigarette smoke exposure could be extended for a longer period of time, these changes in the later-stage model might be more pronounced. In our upcoming research, we will further explore the changes of disease severity and investigate the increases of γδ T cells and M2 macrophages with the animal models of many other longer disease duration-time points. The second limitation is the imbalance between the groups in the proportion of patients in acute exacerbation stage. If the acute exacerbation status of the sampled patients between the two groups was the same, it could be better emphasis the role of periodontal pathogenic bacteria in up-regulating the inflammatory gene expression and inflammatory cytokines content in the lung. And in our forthcoming study, we will further overcome the difficulty of sample collection and obtain the samples without such limitation, to more accurately reflect the difference between the detection of periodontal pathogenic bacteria and the patient’s lung function status.
In summary, we observed that periodontitis could exacerbate COPD progression and identified for the first time that the γδ T-M2 immune mechanism played an important role in mediating periodontitis promoting COPD. The presence of periodontitis intensified the activation of γδ T cells and M2 macrophages in COPD lung tissue, resulting in the aggravation of COPD severity. Therefore, targeting at periodontitis treatment and the γδ T-M2 immune mechanism might be a new practical strategy for the prevention or control of COPD.
MATERIALS AND METHODS
Culture of P. gingivalis
P. gingivalis W83 was grown anaerobically (37°C, 85% N
2, 10% H
2, 5% CO
2). Cells (1 × 10
9 CFU/mL) were collected for following use. For mouse oral infection, the bacteria were centrifuged and resuspended with 2% carboxymethylcellulose (CMC) solution. For the bacteria-cell co-culture experiments, the bacteria were centrifuged and resuspended in 10% fetal bovine serum (FBS) containing DMEM. The bacteria were then co-cultured with cells at a multiplicity of infection (MOI) of 100 (
35,
73).
Experimental animal model
Mouse models were constructed to study the promotion ability and possible immune mechanism that periodontitis affected COPD. The animal model experiment was approved by the animal research committee of West China School of Stomatology, Sichuan University (WCHSIRB-D-2020-127). Male 7-week-old-specific pathogen free (SPF) C57BL/6J mice purchased from Dashuo Biological Technology (Chengdu, China) were randomly divided into several groups (5 mice/group). For periodontitis construction, the 5-0 silk ligatures were tied around the maxillary second molars of mice. Following, mice were infected with P. gingivalis (1 × 109 CFU/mL, 0.2 mL/mice) every other day. Briefly, every other day, at about eight o 'clock in the morning, freshly prepared 1 × 109 CFU/mL P. gingivalis (0.2 mL/mice) were centrifuged and resuspended with 2% carboxymethylcellulose (CMC) solution. And then, the CMC-resuspended P. gingivalis were dipped and transferred onto the disposable oral-specific micro applicators. Finally, mice were orally infected with P. gingivalis by brushing the free gingival area slightly using the bacterial containing disposable oral-specific micro applicators. One week later, COPD models were started to construct using newly purchased cigarettes (Marlboro, 12 mg tar/1.0 mg nicotine; Philip Morris, Richmond, VA) and porcine pancreatic elastase (PPE, RHAWN catalog no. R028727). Mice were exposed to mainstream cigarette smoke (CS) for 2 h every day through the Cigarette Smoke Generators TSE system (TSE Systems China). Two days before CS exposure was begun and on the second day of CS exposure, mice under anesthesia underwent an intratracheal injection of phosphate-buffered saline (PBS) dissolved PPE (2.5 U/mouse each time, in total, 5 U/mouse). For the early-stage COPD model, mice were exposed to CS for 2 weeks, and for the late-stage COPD model, the CS exposure time was extended for an additional 2 weeks. For the anti-γδ T groups (6 mice/group for B group and P group; 9 mice/group for the other four COPD-associated groups), the mice were intraperitoneally injected with γδ-TCR monoclonal antibody (200 ug/mouse; BioXCell catalog no. BE0070) every 2–3 days.
Lung function test
Lung function was measured with the EMMS eSpira Forced Maneuvers system according to the manufacture’s instruction. FEV0.05 value and FEV0.05/FVC value were used to compare the lung function of each group.
H&E staining and methylene blue staining
Five-micrometer paraffin-embedded lung tissue sections were prepared for hematoxylin and eosin (H&E) staining with the H&E staining kit (Solarbio catalog no. G1120) to observe the morphological changes and to analyze disease severity.
The maxillae were stained with 1% methylene blue (Solarbio catalog no. G1303) as described by Wei et al. (
35) and imaged with a microscope. The severity of bone resorption, represented by the distance from the cementoenamel junction (CEJ) to the alveolar bone crest (ABC), was measured to analyze periodontitis severity.
16S rRNA-sequencing
Freshly collected lung tissues were quickly frozen in liquid nitrogen and then transported on dry ice to the Personal Biotechnology company (Shanghai Personal Biotechnology Co., Ltd., China) for 16S RNA-sequencing (V3-V4) on the Illumina platform (Novaseq 6000). The data analysis was carried out with QIIME2 (2019.4).
Tissue DNA extraction and P. gingivalis abundance detection
DNA extraction and
P. gingivalis abundance detection was performed as described previously (
74). Total DNAs of paraffin lung tissues were extracted with the QIAamp DNA Mini Kit (Qiagen, Dusseldorf, Germany).
P. gingivalis abundances of lung tissues were measured through RT-qPCR (described later). The sequences of the
P. gingivalis specific primers were Forward: 5′-
AGGCAGCTTGCCATACTGCG-3′; Reverse: 5′-
ACTGTTAGCAACTACCGATGT-3′.
Immunofluorescence observation
Immunofluorescence staining and observation were performed as previously described (
35). After xylene deparaffinization, gradient alcohol dehydration, hydrogen peroxide solution peroxidation, and goat serum block, the slides containing 5 μm-thick lung tissue slices were incubated with the following primary antibodies, second antibodies, and 49,6-diamidino-2-phenylindole (DAPI): anti-TCR γδ (BioLegend catalog no. 118101), anti-CD206 (Proteintech catalog no. 18704-1-AP), anti-CD86 (Bioss catalog no. bs-1035R), secondary antibodies (BioLegend catalog no. 405510; Proteintech catalog no. SA00013-2), and DAPI (Solarbio catalog no. C0065). Slides that completed the staining procedure were observed and imaged under an Olympus confocal microscope (FV31S-SW V2.4 software).
Flow cytometry
The flow cytometry experiments were carried out according to the procedure outlined by Jin et al., with minor modifications, for analyzing γδ T activating and polarization of macrophage (
29). Briefly, fresh single cell suspensions of lung tissues were firstly stained with the Fixable Viability kit (BioLegend catalog no. L423105 or eBioscience catalog no. 65-0866-18) and then blocked with anti-CD16/32 antibody (BioLegend catalog no. 101320). The cell suspensions were then incubated with the following flowcytometry antibodies (30–60 min): CD45 (BioLegend catalog no. 103139, Elabscience catalog no. E-AB-F1136Q), CD3 (BioLegend catalog no. 100204), F4/80 (BioLegend catalog no. 123130, Elabscience catalog no. E-AB-F0995Q, Elabscience catalog no. E-AB-F0995S), CD206 (BioLegend catalog no. 141706, Elabscience catalog no. E-AB-F1135E), CD86 (BioLegend catalog no. 105008), TCR γδ (BioLegend catalog no. 118118), IL 17 (BioLegend catalog no. 506916, Elabscience catalog no. E-AB-F1272Q), IFN γ (BioLegend catalog no. 113606, Elabscience catalog no. E-AB-F1101D). For intracellular cytokine staining (IL 17 and IFN γ), cells were pre-incubated with Cell Stimulation Cocktail (eBioscience, catalog no. 00-4970-93) for 6 h and Protein Transport Inhibitor Cocktail (eBioscience, catalog no. 00-4980-93) for 2 h (37°C, 5% CO
2) before surface staining. After surface staining, fixation (BioLegend catalog no. 420801), and permeabilization (BioLegend catalog no. 421002), the cell suspension was intracellularly stained. Flow cytometry detection was performed on Attune NxT (Invitrogen Attune NxT flow cytometry software) and Cytoflex (Beckman Coulter Cytoflex flow cytometry software) flow cytometers, and data were analyzed by the FlowJo (V10.8) software and CytExpert software.
Enzyme-linked immunosorbent assay (ELISA)
IL 17 (BioLegend catalog no. 436204) and IFN γ (BioLegend catalog no. 430804) ELISA kits were used to measure the levels of IL 17 and IFN γ in mouse serum samples, lung tissues samples, and the cell culture supernatant samples, respectively.
RT-qPCR
For gene expression levels analysis, total RNA of each lung tissue sample was extracted with RNA extraction kit (Yeasen catalog no. 19221ES50) and reversely transcribed with RNA reverse kit (Yeasen catalog no. 11141ES60). Then, the cDNA samples were used for gene expression level analysis (
Gapdh was used to normalize the expressions of different genes) with the RT-qPCR kit (Syber Green, Yeasen catalog no. 11201ES08). The expression levels were calculated with the 2
−△△CT method (
75). The specific primers for the tested genes were designed using PrimerBanK (
https://pga.mgh.harvard.edu/primerbank/) and synthesized by TSINGKE Biological Technology company (Sichuan, Chengdu), the sequences are listed in Table S3.
For the detection of
P. gingivalis abundance in lung tissue, the gDNA obtained above underwent RT-qPCR procedure.
P. gingivalis abundances were calculated with the 2
−△CT method (
76) and displayed with fold changes to the blank control group.
Ex vivo experiment
The lymphocytes of mouse lung tissues were extracted with the Percoll Cell separation fluid kit (Biosharp catalog no. BS909). BALF were collected with 1 mL sterile PBS and centrifuged to collect the cell precipitate. Lymphocytes or BALF cell precipitate were co-cultured with P. gingivalis in DMEM containing 10% FBS for 24 h.
The peripheral blood mononuclear cells (PBMC) of mice blood were extracted with Peripheral blood lymphocyte separation solution (Solarbio catalog no. P6340). PBMC were co-cultured with P. gingivalis for 24 h. For the anti-γδ T treatment ex vivo part, γδ-TCR monoclonal antibodies (200 ng/mL; BioXCell catalog no. BE0070) were added. Briefly, for the anti-γδ T treatment groups, the inhibitor was first intraperitoneally injected in vivo, and 24 h later, the peripheral blood of mice was collected to obtain the inhibitor-treated PBMC, and then, the obtained PBMC was cultured in vitro. For the corresponding control groups (without γδ-TCR monoclonal antibody treatment), an equal amount of PBS was intraperitoneally injected in vivo, 24 h later, the peripheral blood of mice was collected to obtain PBMC without inhibitor treatment, and then, the obtained PBMC was cultured in vitro.
Flow cytometry procedures were performed and analyzed as described above.
Clinical sample analysis
The clinical samples were collected after the approval and supervision of the Medical Ethics Committee of West China Hospital of Stomatology, Sichuan University (WCHSIRB-D-2022-473). In total, 53 clinical BALF samples from COPD patients were collected at the Clinical Microbiology Lab of West China Hospital. After centrifugation, the precipitates and supernatants were collected and stored separately at −80℃ for future use. The precipitates were used for DNA extraction with the DNA extraction kit (Yeasen catalog no. 18700ES50) and RNA extraction with the same method and kit described above. The obtained gDNA was used for the RT-qPCR with the P. gingivalis specific primers as described above to assess whether the COPD BALF samples had a periodontitis associated state (CT > 35, indicates no detection of P. gingivalis, sample assigned to the COPD-no P. g group; CT < 35, indicates presence of P. gingivalis, sample assigned to the COPD-P. g group). The RNAs were used for gene expression analysis. The specific primers for the tested genes are listed in Table S4. The supernatants were collected for confirmation of IL 17 and IFN γ levels by ELISA with the anti-human IL 17 ELISA kit (MULTI SCIENCES catalog no. EK117HS-96) and IFN γ ELISA kit (MULTI SCIENCES catalog no. EK180HS-96), respectively.
Statistical analysis
Data in this study were represented as the mean ± standard deviation (SD) for at least three independent samples in each group. For the second batch of animal experiments (only two groups: C group and CP group), the ex vivo experiments (except the anti-γδ T treatment portion) and the clinical sample measurement, after a homogeneity test of variance with Levene’s test, t test, or Kruskal-Wallis analysis were used for analysis of differences. For the other experiments, including the multiple-group animal models and four-group ex vivo experiments, one-way ANOVA, and post hoc Tukey’s multiple comparisons were used for analysis of differences among multiple groups, and t test was used for two independent groups. Statistical analysis was performed using GraphPad Prism7 software (version 7.00 for Windows; GraphPad Prism, Inc, La Jolla, CA, USA) with a significance level of 0.05, and all figures were also generated with this software.