1. Introduction
Apical periodontitis is a common oral inflammatory disease primarily caused by invasive anaerobic bacteria from the dental pulp and root canals [
1]. The constant infections eventually lead to the local inflammation and alveolar bone resorption in the periapical region [
2]. Despite root canal treatment (RCT) being performed to remove the infectious substances from the root canal system, nearly 4–15% of teeth still experience AP pain or are even subject to extraction [
3]. Cumulative evidence has put forward the notion that the interplay between pathogens and the host immune system in periapical regions accounts for RCT therapy resistance [
4]. Thus, exploring the causal mechanisms involved in the pathogenesis of AP may be essential for the management of AP.
Recently, Mφ efferocytosis has been reported to influence Mφ polarization [
5,
6,
7]. Mφ efferocytosis is defined as a process where the apoptotic neutrophils in diseased tissues are engulfed by Mφs (defined as efferocytes) [
8]. Notably, after the clearance of apoptotic neutrophils, the efferocytes release various biologically active signaling molecules and extracellular vesicles, which are then taken up by neighboring or distant Mφs to effect phenotypic change, including accelerating Mφs polarized to the M2 phenotype [
5]. The activation of Mφ efferocytosis is composed of three stages [
9,
10]. Firstly, the apoptotic neutrophils in diseased regions spontaneously release the “find me” signals to recruit Mφs. Then, the aggregated Mφs recognize the apoptotic neutrophils via their characteristic cell surface changes. The most familiar cell surface change is the exposure of phosphatidylserine (PS), a critical “eat me” signal, on the surface of dying cells [
11]. In addition, MerTk (C-mer proto-oncogene tyrosine kinase), the key receptor on the surface of Mφs, can recognize and bind to PS [
11]. Moreover, Gas6, a bridging protein, is responsible for bridging PS indirectly with efferocytosis-related receptors on Mφs’ surface [
12]. Once finishing the binding step, the apoptotic neutrophils start to be swallowed and degraded by the efferocytes, which can accelerate the polarization of Mφs toward a pro-resolving M2 phenotype [
5].
In view of the crucial role of Mφ efferocytosis on apoptotic cells’ clearance and Mφ polarization, it may not be surprising that Mφ efferocytosis is involved in the etiology and pathogenesis of multiple inflammatory diseases such as atherosclerosis (AS), osteoarthritis (OA) and periodontitis [
13,
14,
15,
16]. Lv et al. demonstrated that increasing the Mφ efferocytosis ability in an AS model in ApoE
−/− mice could effectively suppress AS progression, even leading to the resolution of AS [
17]. Moreover, Sordo et al. found that there was a marked reduction in the fraction of synovial tissue Mφs engaging in efferocytosis in patients with late-stage knee OA, resulting in the accumulation of apoptotic synovial cells and then exacerbating the disease activity in OA patients [
18]. In order to further understand the role of efferocytosis in OA, Yao et al. increased the Mφ efferocytosis ability in an OA mice model via the intra-articular injection of Gas6, and then confirmed that targeting Mφ-associated efferocytosis could significantly suppress OA progression [
14]. Particularly, research given by Kourtzelis et al. and Li et al. delineated the relation between efferocytosis and periodontitis [
15,
16]. They unveiled that Mφ efferocytosis participates in the progression of periodontitis. The regulation of Mφ efferocytosis is closely correlated with inflammation resolution in periodontal tissues [
15,
16]. Taken above, Mφ efferocytosis highlights great capacities to suppress the deterioration of inflammatory diseases or accelerate the resolution of inflammation. However, the role of Mφ efferocytosis in the pathogenesis of AP is still elusive. Therefore, this study aims to outline the evidence for the putative involvement of macrophage efferocytosis in the pathogenesis of AP.
In the current study, we used human periapical tissues as well as a cell model to investigate the involvement of Mφ efferocytosis in AP. Next, we constructed a co-cultured cell model and a mouse model of AP, with the application of efferocytosis agonist (ARA290), to explore the role of Mφ efferocytosis in attenuating AP progression. Deeper investigations and understanding into the role of efferocytosis in AP can provide therapeutic targets for inflammation resolution and tissue regeneration.
3. Discussion
Emerging evidence has been displayed regarding the effect of Mφ efferocytosis in suppressing inflammation progression and accelerating inflammation resolution [
5,
6,
7]. In the current study, we discovered that Mφ efferocytosis participates in the AP pathology and promotes the resolution of AP by accelerating M2 Mφ polarization. Moreover, strengthening the capacity of Mφ efferocytosis could significantly ameliorate the inflammation status and bone loss in AP in in vitro and in vivo models. Notably, this study has initially observed the emergence of Mφ efferocytosis in human periapical tissues, further suggesting Mφ efferocytosis may be involved in AP pathogenesis.
Mφ efferocytosis represents the process of the timely and effective removal of undesirable cells, such as apoptotic neutrophils, by efferocytes [
5]. This specialized process is of great importance for essential body functions, including immunoregulation, organism growth and the maintenance of tissue homeostasis [
7]. During the valid process of Mφ efferocytosis, apoptotic neutrophils are cleared as they should be to block their harmful effects via avoiding their leakage to normal tissue microenvironments; or else, apoptotic neutrophils can be apt to secondary necrosis, leading to the release of detrimental autoantigens into healthy tissues [
19]. Crucially, studies have elucidated that the impairment in the efferocytic course postponed inflammation resolution and aggravated the pathology in various inflammatory diseases, including neurodegenerative disorders, atherosclerosis, osteoarthritis and the like [
18,
20,
21]. It is well known that Mφ efferocytosis facilitates the polarization of Mφs to an M2 phenotype, which stimulates the Mφ efferocytotic capacity in a feedback loop [
22]. Cai et al. clarified that enhancing the efferocytosis efficiency of Pg.LPS-stimulated J774a.1 Mφs could markedly down-regulate the expression of proinflammatory cytokine TNF-α, whereas the expression of anti-inflammatory cytokine IL-10 was significantly up-regulated. Notably, the M1/M2 Mφ ration in the above cell model has also been down-regulated simultaneously, which suggested the regulatory effect of Mφ efferocytosis on Mφ polarization [
23]. Moreover, Bhattacharya et al. further confirmed the role of efferocytosis on Mφ phenotype remodeling [
5]. They depicted that after the engulfment of apoptotic neutrophils, the efferocytes secrete soluble mediators or extracellular vesicles, which can affect the Mφ phenotype remodeling in an autocrine/paracrine manner, promoting the polarization of Mφs toward a pro-resolving M2 phenotype [
5]. In this study, we found that, in AP patients and the co-cultured cell model, as the AP progresses, the efferocytotic ability decreased (
Figure 1 and
Figure 2). Whereas, in comparison with the healthy periapical tissues, the protein levels of Gas6 and Mertk increased significantly in the AP clinic samples, including PGs and RCs. This may be attributed to the number of apoptotic neutrophils far outpacing the number of available efferocytes, leading to the periapical tissue suffering from continuous damage (
Figure 2). Notably, as the efferocytosis rate increased in the inflamed co-cultured cell model, the expression of the anti-inflammatory cytokine (IL-10) up-regulated in parallel; however, the expression of the proinflammatory cytokine (IL-1β) and the ration of M1/M2 decreased correspondingly (
Figure 4). This is particularly intriguing and raises the possibility that enhancing the Mφ efferocytosis could suppress the inflammation status and accelerate inflammation resolution in periapical lesions.
Previous data elucidated that ARA290 can promote the efferocytosis of Mφs in vitro, and the application of ARA290 enhances apoptotic cell uptake [
24]. ARA290 is an erythropoietin-derived helix-B peptide and has been reported to retain the anti-inflammatory and tissue-protective functions of erythropoietin. Huang et al. claimed that ARA290 could reduce the serum concentrations of inflammatory cytokines IL-6, MCP-1 and TNF-α in systemic lupus erythematosus (SLE) mice [
24]. Further, ARA290 suppressed the inflammatory activation of Mφs and promoted the uptake of apoptotic cells by Mφs, thus ameliorating SLE clinical and pathological manifestations [
24]. Moreover, a study conducted by Xu et al. further validated the anti-inflammatory effect of ARA290. Daily administration of ARA290 could ameliorate depression-like behavior during chronic stress induction in mouse models via reducing neutrophils in the bone marrow and meninges [
25]. Here, we tested the efferocytosis ability of Pg. LPS+IFN-γ-stimulated dTHP-1 with different concentrations of ARA290. According to our results, the expression of efferocytosis markers accelerated after treatment with ARA290 in a dose-dependent manner under the Pg.LPS + IFN-γ challenge (
Figure 5). Moreover, the M1/M2 ration significantly decreased with the treatment of ARA290 under the 300 nM and 600 nM concentrations (
Figure 5). Subsequently, we detected the role of ARA290-enhanced Mφ efferocytosis in regulating AP pathogenesis and uncovered that intraperitoneally administration restrained the inflammatory status and bone loss in periapical lesions (
Figure 6). The promoting effect of Mφ efferocytosis on M2 Mφ polarization has been stressed before [
26]. In consistency with a previous study, we found that ARA290 could accelerate the polarization of Mφs to the M2 phenotype and trigger the production of anti-inflammatory cytokines via enhancing the Mφ efferocytotic capacity in periapical lesions. Notably, a weak co-localization of Gas6 and c-caspase-3 was found in the ARA290-treated AP animal model when compared with the AP group; thus, we speculated that how ARA290 exerts its biological functions mainly depends on the Mertk signals. Moreover, in the ARA290-treated co-cultured cell model, we only detected the expression of Mertk, as the efferocytosis of THP-1 macrophages could not be transducted via Gas6 [
27]. The above results imply that targeting Mφ efferocytosis reduces the inflammatory status and suppresses bone loss in the periapical region.
Considering the facilitating effect of Mφ efferocytosis on inflammation resolution, growing evidence has emerged to explore the biomarker that can target Mφ efferocytosis. Meriwether et al. revealed that Mφ cyclooxygenase 2 (COX2) could accelerate intestinal epithelial repair by mediating Mφ efferocytosis and efferocytosis-dependent reprogramming [
28]. Moreover, developmental endothelial locus-1 (DEL-1) has also been demonstrated to regulate efferocytosis-induced Mφ reprogramming to a pro-resolving phenotype, promoting the resolution of experimental periodontitis [
16]. Furthermore, in stroke mice, STAT6 activation in microglia and Mφs improved efferocytosis and modulated microglia/Mφ phenotype, accelerated inflammation resolution and ameliorated stroke outcomes [
6]. This study has some limitations. In this study, we only focused on the involvement of Mφ efferocytosis in AP pathogenesis. Whereas more studies are being conducted by our research group, which aim at exploring the effective biomarkers that are capable of enhancing the Mφ efferocytosis capacity in AP. Moreover, the identification of efferocytes in in vitro or in vivo models mainly depends on indirect testing, including flow cytometry, IHC staining, IF staining, etc. In the future study, direct testing, such as electron microscopy (EM), should be conducted to observe the efferocytes in periapical lesions.
4. Materials and Methods
4.1. Clinic Samples Collection
The clinic apical periodontitis samples were collected from 18 subjects who were diagnosed with AP according to clinical and radiographic examination during endodontic microsurgery. Clinical manifestation covers a painful response to palpation or percussion or biting, no response to pulp vitality tests, fistula and sinus tract. In addition, the X-ray presented an apical radiolucency. Notably, the subjects were without any symptoms of acute AP. Healthy periapical tissues were obtained from 16 subjects who underwent permanent tooth extraction for orthodontic purposes. All subjects who enrolled in this study were free of systemic diseases and did not accept antibiotic treatment during the last 6 months. The collected tissues were fixed in 4% paraformaldehyde for 24 h and then embedded in paraffin. After that, the samples were consecutively sectioned into 4 μm slides for later experiments.
4.2. Induction of Mice AP Model and ARA290 Administration
In total, thirty male C57BL/6 mice with a body weight of 20–25 g (aged 6–8 w) were randomly assigned into 3 groups (
n = 10/group): Control (Cont) group, apical periodontitis (AP) group and AP+ARA290 group. The mice were anesthetized with ketamine hydrochloride 10% (150 mg/kg body weight) and xylazine 2% (7.5 mg/kg body weight). Then, the bilateral first molar of the mice mandibles was perforated using a high speed 1/4# round bur until the pulp chamber was exposed [
29]. Following the pulp operation, ARA290 (120 μg/kg, MCE, Monmouth Junction, NJ, USA) was intraperitoneally injected into mice three times per week [
24]. Four weeks after AP induction, the mandible was isolated and fixed in 4% paraformaldehyde. The left side of the mandible was separated for micro-CT scanning and histology. The right hemimandibles were extracted for further immune testing. In this study, thirty C57BL/6 mice were kept in a regulated enviroment at a temperature of 25 °C, with access to food and water at all times and a 12 h light and dark cycle. All of the mice were managed sustainably to avoid mental and physical disorders.
4.3. Cell Culture and Treatments
Human monocytic leukemia THP-1 cells were purchased from Procell Life Science & Technology Co., Ltd. (Wuhan, China). Human promyelocytic leukemia cells (HL-60) were gifted from the Department of Hematology, the First Affiliated Hospical of Xi’an Jiaotong University (Xi’an, China). The above two cell lines were incubated in PRMI-1640 medium (Gibico, Billings, MT, USA) supplemented with 10% FBS (BI, Kibbutz, Beit Haemek, Israel) and cultured at 37 °C with 5% CO
2 and 95% relative humidity. THP-1 cells were primed with 100 ng/mL phorbol 12-mysistate 12-acetate (PMA, Sigma-Aldrich, St. Louis, MO, USA) for 24 h to differentiate into Mφ (dTHP-1) [
30]. Amounts of 100 ng/mL P. gingivalis lipopolysaccharide (Pg.LPS, Sigma-Aldrich, St. Louis, MO, USA) and 40 ng/mL interferon-γ (IFN-γ, Santa Clara, CA, USA) were then added into dTHP-1 to mimic the inflammatory conditions in the infected periapical region [
31,
32].
HL-60 cells were treated with 1.3% DMSO (Sigma-Aldrich, USA) for different time points; in detail, 1 d, 2 d, 3 d, 4 d and 5 d to differentiate into a neutrophil-like granulocyte (dHL-60). Western blot analysis was conducted to detect whether HL-60 was successfully differentiated into dHL-60. Next, in order to induce cell apoptosis, dHL-60 cells were further treated with 100 nM PMA for 24 h [
33]. Flow cytometry was performed to confirm the apoptosis of dHL-60.
To investigate the phagocytic efficiency of Mφ on apoptotic cells under AP conditions, the Pg. LPS+IFN-γ-stimulated dTHP-1 cells were co-cultured with apoptotic dHL-60 cells, at a ratio of 1:10 for 0.5 h, 1 h, 3 h, 6 h and 12 h. To further explore the role of Mφ efferocytosis in AP pathology, the co-cultured cell model was simultaneously treated with ARA290 (#269143, MCE, Monmouth Junction, NJ, USA), a well-known efferocytosis agonist, under the following different ARA290 concentrations: 100 nM, 300 nM and 600 nM for 24 h.
4.4. Flow Cytometry
The rates of dHL-60 cell apoptosis were detected via flow cytometry (FCM) analysis using an Annexin V-FITC/PI Apoptosis kit (Bestbio, Nanjin, China). dHL-60 cells were seeded in a 6-well plate with 100 nM PMA for 24 h, then re-suspended in 400 μL binding buffer and stained with 10 μL of PI (propidium iodide) and 5 μL of FITC Annexin V. The treatments were continued for 16 min at 4 °C in the dark conditions. After that, the samples were submitted to a flow cytometer (Agilent, BD FACSCalibur, Santa Clara, CA, USA). Annexin V-positive and PI-negative dHL-60 cells were determined as the early apoptotic cells; furthermore, Annexin V- and PI-positive cells were identified as the late apoptotic cells.
The results were analyzed by FlowJo 1.6.0 software (Ashland, OR, USA).
4.5. Macrophage Efferocytosis Testing
To precisely explore the role of Mφ efferocytosis in the progression of AP, dHL-60 cells were firstly marked with fluorescence CFDA SE (Beyotime Biotechnology, Shanghai, China). Fluorescent dHL-60 cells were induced into apoptosis and then, the apoptotic dHL-60 cells were overlaid on the dTHP-1 (10:1) for 0.5 h, 1 h, 3 h, 6 h and 12 h at 37 °C under inflammatory conditions. Following efferocytosis, cells were rigorously washed three times with 1× ice-cold PBS to remove the dHL-60 cells that were not being engulfed. After that, the cells were fixed with 0.5% paraformaldehyde and incubated with CD68 antibody (1:100 dilution, Biolegend, San Diego, CA, USA) for 2 h at 37 °C to label dTHP-1. The efferocytotic efficiency was observed and analyzed under a confocal microscope (Olympus, Tokyo, Japan) at 400× magnification. Mφ efferocytosis was determined by counting cells including phagocytic green fluorescent apoptotic bodies. Data were represented as percent (%) efferocytosis–total number of cells with ingested apoptotic dHL-60 cells divided by the total number of dTHP-1 counted times 100.
Furthermore, flow cytometry was conducted for detecting the Mφ efferocytosis during the AP course. In brief, the apoptotic dHL-60 cells were marked with PKH26 (MCE, Monmouth Junction, NJ, USA) and were subsequently added to dTHP-1 cells under inflammatory conditions for different times as follows: 30 min, 1 h, 3 h, 6 h and 12 h. After being fixed with 0.5% paraformaldehyde, the co-cultured cell model was incubated with CD68-FITC antibody (1:100 dilution, Biolegend, Hangzhou, China). Then, the efferocytosis activity was measured via flow cytometry (BD FACSCalibur, CA, USA). Histograms were plotted using FlowJoTM version 10 software (BD Biosciences, San Jose, CA, USA).
4.6. Micro-Ct
Fixed left hemimandibles were scanned as previously described using a cone beam-type tomograph (QuantumGX, PerkinElmer, Hopkinton, MA, USA). Scanning parameters were set as follows: voltage, 90 kV; current, 88 μA; FOV, 25 mm; voxel size, 50 μm. The scanned data were reconstructed and analyzed with Mimics 17.0 software (Materialize, Leuven, Belgium). Briefly, the lesion size was acquired via the subtraction of an averaged normal periodontal space in baseline controls from a total periapical radiolucent area and expressed as square millimeters. After micro-ct, the specimens were submitted to hematoxylin–eosin staining.
4.7. Hematoxylin–Eosin Staining
The decalcified left mandibles were embedded in paraffin and sectioned longitudinally at 4 μm thickness in mesio-distal orientation according to a general histology protocol. The slides were then dewaxed in xylene and rehydrated in gradient alcohols. Hematoxylin and eosin (HE) staining was performed on consecutive tissue sections. The slices with apical foramen were stained with HE according to the manufacturer’s manual (Solarbio, Beijing, China). Images were photographed and observed under light microscopy under 4× magnification (Olympus, Tokyo, Japan).
4.8. Immunohistochemical (IHC) or Immunofluorescence (IF) Staining
IHC and IF staining were conducted as a previous study described [
34]. In brief, IHC staining was carried out by the Streptavidin–Biontin Complex (SABC) method on the basis of the manufacturer’s protocol (Boster, Wuhan, China). The sections were treated with digestive solution (Boster, Wuhan, China) to remove the antigen at 37 °C for 25 min. Thereafter, 3% hydrogen peroxide was added to the samples for endogenous peroxidase retrieval. Samples were then blocked with 5% BSA and incubated with the corresponding primary antibodies: anti-IL-1β (1:100 dilution, Santa Cruz, CA, USA), IL-10 (1:100 dilution, Santa Cruz, CA, USA), CD11b (1:100 dilution, Proteintech, Wuhan, China), c-caspase-3 (1:150 dilution, Bioss, Beijing, China), Mertk (1:100 dilution, Bioss, Beijing, China), Gas6 (1:100 dilution, Bioss, Beijing, China), CD68 (1:150 dilution, Bioss, Beijing, China), CD86 (1:150 dilution, Bioss, Beijing, China), CD163 (1:150 dilution, Bioss, Beijing, China). After 18 h, the slides were treated with secondary antibodies (Zhongshanjinqiao, Beijing, China) for 1.5 h. The immune reaction was observed with DAB substrate kit (Boster, Wuhan, China).
For the IF staining, after being blocked with 5% BSA, the tissue samples and the cell samples were incubated with the primary antibodies against CD11b and c-caspase-3, Mertk and CD68, Gas6 and CD68, Mertk and CD86, Mertk and CD163, Mertk and c-caspase3, Gas6 and c-caspase3. Of note, the dilution of the above primary antibodies was 1:150. Then, the secondary antibodies (dilution with 1:150, Boster, Wuhan, China) were goat against rabbit CY3 or goat against mouse FITC. IHC images were obtained using the Nikon microscope image system (Nikon Ltd., Tokyo, Japan). The immunology-positive cells were inspected and captured using a confocal microscope (Olympus, Tokyo, Japan) at 40× magnification. Semi-quantitative analysis was conducted using Image J software (Version 8, NIH, Bethesda, MD, USA). All of the experimental steps were taken by two independents in a double-blind manner.
4.9. Western Blot Analysis
All experiments were conducted in at least triplicates. The dTHP-1 cells were seeded at 5 × 104 /well in 6-well plates with the stimulation of Pg.LPS and IFN-γ. Then, the apoptotic dHL-60 cells were added to the above cells to establish the co-cultured cell model. At the time points 0.5 h, 1 h, 3 h, 6 h and 12 h, after the co-cultured cell model was established, 150 μL RIPA lysis buffer containing 1 mM phenylmethanesulfonyl fluoride was added to the cells to obtain the cell lysate. Then, the supernatant was harvested after centrifuging at 12,000× g for 10 min at 4 °C. Thereafter, the supernatant was mixed with lading buffer (Boster, China) and boiled at 100 °C for 5 min. Equal amounts of protein (25 μg) were loaded on SDS-PAGE and electrophoresed at 60 V for 40 min, and then the voltage was switched to 110 V for 1 h. Then, the proteins were transferred onto polyvinylidene fluoride (PVDF) membranes (Millipore, St. Louis, MI, USA). The blots were then covered with primary antibodies against Mertk (1:300 dilution), IL-1β (1:300 dilution), IL-10 (1:300 dilution), β-actin (1:3000 dilution) at 4 °C for 18 h, followed by incubation with peroxidase-conjugated secondary antibodies. The immune blots were observed with enhanced chemiluminescence detection reagent (Millipore, St. Louis, MI, USA). Β-actin was set as the internal controls for the total protein. Image J software (Version 8, NIH, Bethesda, MD, USA)was operated to analyze the densitometry of the bands from the Western blot experiment. Notably, all of the primary antibodies were purchased from Bioss, China.
4.10. Reverse Transcription Quantitative Polymerase Chain Reaction (RT-qPCR)
Total RNA was extracted from the co-cultured cell model according to the TRIzol reagent protocol (Thermo Fisher Scientific, Waltham, MA, USA). The purity and the concentration of RNA was detected in a NanoDrop. Complementary dna (cDAN) was synthesized using RevertAid First Strand cDNA synthesis kit (Thermo Fisher Scientific, MA, USA), according to the manufacturer’s instructions. SYBR-green-based RT-PCR was performed with 50 cycles of 94 °C (1 min), 56 °C (1 min) and 72 °C (2 min) on an ABI PRISM 7700 (Applied Biosystems, Woburn, MA, USA). The analysis was conducted using the 2
−△△CT method. The primer sets used in this study are shown in
Table 1.
4.11. Statistical Analysis
Statistical analysis was conducted using GraphPad Prism 8.0 software (GraphPad Software, Inc., La Jolla, CA, USA). All numerical data are presented as mean standard deviation. Multiple group comparisons were performed using a one-way analysis of variance (one-way ANOVA). p < 0.05 was considered statistically significant.