1. Introduction
Lidocaine, a commonly used local anesthetic, also has anti-inflammatory activity in various diseases. Lidocaine also regulates the immune system [
1]. Although lidocaine inhibits natural killer (NK) cell function at high concentrations, it stimulates the killing activity of these cells at therapeutic plasma concentrations [
2]. Ramirez and colleagues demonstrated that lidocaine enhances NK cell killing activity against three different leukemia cell lines [
3]. In the past, there were many reports on the regulation of immune cells by lidocaine, the production of inflammatory cytokines, and the regulation of nuclear factor-kappaB (NF-κB) [
4,
5,
6]. It was found that lidocaine downregulates NF-κB signaling and inhibits cytokine production and T cell proliferation [
6,
7]. Uncontrolled immune responses are associated with almost all kinds of cancer. Immune cells play a crucial role in anti-cancer responses and cytokine production. Therefore, inhibition of the activation of regulatory T (Treg) cells and their related cytokine production is considered an important strategy to deal with cancer [
8]. Treg cells are a subset of CD4
+CD25
+ T cells that potently suppress many immune responses [
9].
IFN-γ, a cytokine essential for both innate and adaptive immune responses, is produced principally by CD4
+ and CD8
+ T cells. It is critical for successful clearance of intracellular pathogens and also in host defense against malignant transformation [
10]. IFN-γ production should therefore be subject to intense positive and negative regulation in cells of the immune system [
11]. The specific immunity is controlled by specific cytokines [
12]. Most of the IL-12-induced effects are mediated by IFN-γ [
13]. IL-12 induces immune responses against tumors through their direct effects on tumors via angiogenesis and lymphocytes [
13]. CD14
+ macrophages commonly exist in two distinct subsets: classically activated (M1) macrophages, which are pro-inflammatory and associated with T-helper (Th)1 cytokines such as IFN-γ and IL-12, and alternatively activated (M2) macrophages, characterized by markers such as TGF-β and IL-10 [
14]. M2 macrophages secrete high amounts of IL-10 and TGF-β to suppress the inflammation [
14]. IL-10 is an important anti-inflammatory cytokine produced under different conditions of immune activation by a variety of cell types, including T cells and macrophages [
15]. It seems to be a double-edged sword in the host defense.
IL-35 is secreted by forkhead box P3 (Foxp3)
+ cluster of differentiation CD4
+CD25
+ Tregs or a Foxp3
-Treg population and has previously been proposed as a novel immune-suppressing cytokine and a key effector molecule of Tregs function [
16]. It shares the IL-12 p35 and Epstein-Barr virus-induced gene 3 subunits with IL-12 and IL-27, respectively [
17]. IL-35 suppresses the activity of Th1, Th2 and Th17 cells, and expands CD4
+CD25
+Foxp3
+Tregs [
16]. It is also required for maximal Treg activity and it alone is sufficient to suppress T-cell proliferation [
18]. Moreover, IL-35 stimulation increased the inhibitory function of CD4
+CD25
+Foxp3
+ Tregs and enhances IL-35/IL-10 productions [
18]. However, the precise underlying mechanism behind the involvement of IL-35 in lidocaine effects has yet to be elucidated in normal human PBMCs and gastric TIICs.
Accumulating evidence indicates that CD4
+CD25
+Treg cells have a powerful ability to suppress the host immune response [
8]. However, recent studies have shown that tumor cells can recruit these Treg cells to suppress antitumor immunity in the tumor microenvironment (TME), thereby limiting the efficiency of cancer immunotherapy [
9]. Our strategy is to use lidocaine to overcome tumor-associated immunosuppression for successful gastric cancer immunotherapy. It was reported that PD-1 play a vital role in inhibiting immune responses [
19,
20]. Monoclonal antibodies target PD-1 can boost the immune response against cancer cells [
19,
20]. PD-1 blockades have also been associated with the development of cytotoxic CD8
+ T cells and an imbalance of the immune system [
20,
21]. This is the first report to reveal that lidocaine blockade of PD-1 and increase IFN-γ through NF-κB signaling. Lidocaine-treated gastric CD8
+ TIICs augmented the anti-tumor response, killing primary gastric cancer cells (PGCCs).
2. Materials and Methods
2.1. Isolation and Culture of PBMCs from Healthy Adult Volunteers
Blood samples were obtained from healthy adult volunteers (participants without known medical conditions) (n=23) at Taichung Veterans General Hospital, following institutional review board (IRB) approval and consent. The protocol was approved by the Ethical and Scientific Committee of Taichung Veterans General Hospital (IRB no. SF22141B#1). Peripheral blood was collected in EDTA tubes, and peripheral blood mononuclear cells (PBMCs) were isolated using Histopaque 1.077 g/ml (Sigma Chemicals, Darmstadt, Germany), as previously described [
22]. The cell pellet (PBMCs) was resuspended in RPMI medium supplemented with 10% fetal bovine serum (FBS), and 100 U/ml penicillin and streptomycin.
2.2. Reagents and Antibodies
Reagents and sources were as follows: FITC-conjugated IgG1 anti-human CD14 (BD Bioscience, San Diego, USA); FITC-conjugated IgG1 anti-human CD8 (eBioscience); FITC-conjugated IgG1 anti-human CD25 (Elabscience); IgG1-FITC isotype control (Caltag Laboratories, Inc., Burlingame, USA); PE-conjugated IgG1 anti-human CD4 (Elabscience, San Diego, USA); PE-conjugated IgG1 anti-human IFN-γ (Elabscience, San Diego, USA); IgG1-PE isotype control (Caltag Laboratories, Inc., Burlingame, USA). NF-κB-specific inhibitor BAY11-7082 was purchased from Merck (San Diego, USA).
2.3. Isolation of TIICs from Gastric Cancer Patients
Ex vivo malignant gastric tissues were obtained from patients undergoing routine planned cancer-related surgery (n=21). Written informed consent was obtained from each patient in accordance with local institutional ethics review and approved by the Ethical and Scientific Committee of Taichung Veterans General Hospital (TCVGH-IRB no. SF22141B#1). The generation of TIIC cultures from tumors has been detailed elsewhere [
23]. In brief, tumor specimens removed from cancer patients were placed on a plate with 5% FBS in Hank’s balanced salt solution buffer (Gibco, New York, USA) on ice and disintegrated using scissors. The homogenate was collected and treated with 1 mg/ml type IV collagenase (Sigma, St. Louis, USA) and 0.05 mg/ml DNase (Promega, Madison, USA) for 30 minutes at 37°C with gentle agitation. The digested extract was screened using a 100-mesh filter, and the filtrate was washed with 5% FBS in Hank’s balanced salt solution buffer and centrifuged at 600xg for 7 minutes at 4°C. The cell pellet obtained was treated with ACK erythrocyte lysis buffer (155 mM NH
4Cl, 10 mM KHCO3, and 1 mM Na
2EDTA, pH 7.3) for 5 min at room temperature. Finally, TIICs were resuspended in RPMI 1640 medium with 10% FBS. Cells were harvested by 7 to 14 days of culture. Each initial well was considered to be an independent TIICs culture and was maintained separately from the others.
2.4. Primary Gastric Cancer Cells (PGCCs) Culture from Fresh Surgical Malignant Gastric Tissues
Specimens were collected in Dulbecco’s modified Eagle’s medium (Biochrom co, Berlin, Germany) containing 1% Penicillin/Streptomycin for transport to our laboratory. Primary PGCCs were purified and maintained in Dulbecco’s modified Eagle’s medium supplemented with 10% FBS as previously described [
24,
25]. On the next day, the cell culture was rinsed with PBS twice to remove non-adherent cells. The medium was changed every 3-7 days, depending on the density of cell growth. The colonies increased in size and spread out, resulting in some cells separating at the periphery of the colonies after 2 weeks of culture. PGCCs were identified using granulin (GRN) markers [
24]. These primary cells were maintained in culture for up to 4-8 weeks.
2.5. Sorting of CD8+T cells, CD4+CD25+ Treg, CD14+Macrophages, and GRN+ PGCCs
PBMCs were obtained from healthy donors, TIICs and PGCCs were obtained from gastric cancer patients after written informed consent and approval by the Ethical and Scientific Committee of Taichung Veterans General Hospital (IRB no. SF22141B#1). Cells were stained with the CD8 Ab for cytotoxic T cells (CTLs), CD4 Ab and CD25 Ab for Tregs, CD14 Ab for macrophage cells. PGCCs were identified using GRN markers [
24]. Sample acquisition and cell sorting were managed on the BD FACSMelody™ cell sorter (BD Biosciences, San Jose, USA) and analyzed using BD Chorus software.
2.6. Human CD8+, CD4+CD25+, CD14+PBMCs and CD8+, CD4+CD25+, CD14+ TIICs Viability
Human immune cells were assessed by flow cytometric analysis using propidium iodide-stained cells. Initially, 104 cells were incubated in 96-well plates in the absence or presence of lidocaine at concentrations ranging from 0.25 mM to 1.5 mM. After 72h treatment, cells were washed with PBS and fixed with 70% ethanol for 1 h on ice. Pelleted cells were then incubated with RNase-A (0.1 mg/mL) and propidium iodide (40 μg/mL) for 1 h with shaking and protected from light. The percentage of the sub-G1 population was determined by flow cytometry.
2.7. CD8+ Primary T Cells and CD14+ Primary Macrophage Activation
CD8+PBMCs and CD14+PBMCs were triggered by phorbol myristate acetate (PHA) 2.5 mg/ml and phytohaemagglutinin (PMA) 50 ng/ml in the absence or presence of lidocaine concentrations ranging from 0.25 mM to 1.5 mM, or cultured in media alone as a negative control.
2.8. Analysis of Cytokines by ELISA
Human IFN-γ, IL-12, IL-10, TGF-β, and IL-35 protein levels were quantified using a sandwich ELISA. The production of IFN-γ, IL-12, IL-10 and TGF-β were analyzed by human IL-10 ELISA kit (Arigo Biolaboratories, Taipei, Taiwan), human IL-12, TGF-β ELISA kit (Elabscience, Houston, USA), and human IFN-γ ELISA KIT (Gen-Probe, San Diego, USA) respectively. Initially, 100 μL of supernatant was added to the ELISA plates with pre-coated monoclonal antibody at 37 °C for 2 h. After washing with PBS three times, the plates were blocked with complete RPMI medium containing 10% fetal bovine serum (FBS) for 2 h at 37 °C. Finally, 100 μL of streptavidin-HRP was added to each well for 20 min at room temperature. After washing 3 times, 100 μL of 3, 3′,5′,5′-tetramethylbenzidine (TMB) coloring agent was added to each well. Finally, color development was initiated by adding 100 μL of TMB buffer (100 μL/well) and terminated by adding 100 μL of H
2SO
4. The OD
450 nm value was measured with an ELISA reader [
22]. IL-35 concentration was assayed using a human IL-35 ELISA kit (#88-7357; eBioscience Inc., San Diego, USA) according to the manufacturer’s protocol. The optical density was measured at 450 nm. The reported concentration of IL-35 was determined by subtracting the concentration of IL-35 in PBS alone and represented by relative units compared to standard samples provided in the kit. All assays were performed in triplicate.
2.9. Foxp3 Staining and Intracellular Cytokine Staining
CD4
+CD25
+ Tregs showed Foxp3 expression as determined by FACS intra-cytoplasmic staining with APC-conjugated IgG1 anti-Foxp3Ab (eBioscience, San Diego, USA). For IL-10, TGF-β, and IL-35 detection, PE-conjugated IgG1 fluorescent antibodies were utilized (all from BD Pharmingen, San Diego, USA). We used the Intracellular Cytokine Staining Kit (BD biosciences) consisting of fixation/permeabilization buffer. In brief, the cells were fixed using fixation/permeabilization buffer, followed by washing the cells with PBS and then staining the cells using anti–Foxp3 and anti–IL-10, TGF-β, and IL-35 antibodies. The cells were finally suspended in 0.5 ml staining buffer and analyzed as previously described [
26].
2.10. Detection of PD-1+ Analysis of TIICs by Flow Cytometry
After washing three times with PBS buffer; 10
4 cells of TIICs were divided into 1.5 cc centrifuge tubes. Fluorescent-conjugated anti-PD-1 antibodies; along with their corresponding fluorochrome-conjugated mouse immunoglobulin G isotype controls; were all obtained from PharMingen (San Diego; USA). Antibodies were added; and the reaction was carried out in the dark at 4°C for 30 min. After washing once; the cells were resuspended in PBS and analyzed by flow cytometry using BD FACSCalibur (Becton Dickinson; USA); as previously described [
22]. Representative results are shown in histograms based on 10
4 gated cells in all conditions, and cell viability was >95%, as assessed by propidium iodide exclusion. Similar results were observed using at least 3 different TIICs donors.
2.11. Apoptosis Assays
Apoptosis was assessed by flow cytometric analysis of cells stained with annexin V-fluorescein isothiocyanate (FITC) and 7-AAD according to manufacturer’s instruction (Annexin V-FITC/7-AAD Apoptosis Kit, Elabscience, Houston, USA). TIICs and PGGCs co-culture cells were seeded at 106 cells/mL in 6-well plates followed by treated with lidocaine (1.5mM) or medium only for 72h. Briefly, cells were washed and resuspended in 500 μL PBS followed incubation with 10 μL 7-AAD and 10 μL annexin V-FITC at room temperature in the dark for 15 min and analyzed by flow cytometry.
2.12. Detection of Cytotoxicity of CD8+TIICs and PGCCs
The cytotoxicity of CD8
+TIICs and PGCCs were estimated by quantification of LDH activity in the culture medium by using the QuantiChrom
TM LDH Cytotoxicity Assay Kit (BioAssay Systems, Hayward, USA) [
27,
28]. Briefly, cytotoxicity assays were carried out in 96-well plates with a final sample volume of 100 μl/well. Target cells (PGCCs) in 50 μl/well were co-cultured with effector cells (CD8
+TIICs) at various effector to target ratios (5:1) for 4h [
29].
2.13. Lidocaine Treated-CD8+TIICs Mediated Cytotoxicity Assay Using Time-Resolved Fluorometry
Lidocaine (1.5 mM) treated CD8
+TIICs mediated cytotoxicity was determined using the DELFIA
® EuTDA Cytotoxicity Reagents (PerkinElmer Life Sciences, Waltham, MA, USA), as described previously [
30,
31,
32]. Briefly, target cells (PGGCs) were incubated with freshly prepared 10 μM BATDA (a fluorescence enhancing ligand) in 2 ml of culture medium for 30 min at 37°C, and washed. Next, 100 μl of BATDA-labeled target cells (PGCCs) were transferred into a round bottom sterile plate and co-cultured with lidocaine (1.5 mM) treated CD8
+TIICs for 2 hours at effector/target ratios was 5:1. After incubation, 20 μl of supernatant from each well was transferred to the wells of flat-bottom 96 well plates. 180 μl of europium (Eu) solution was then added to form highly fluorescent and stable chelates (EuTDA), and the fluorescence of these chelates were measured by time resolved fluorometry (Enspire 2300-0000, PerkinElmer). The percent of specific release was calculated using (experimental release – spontaneous release)/ (maximum release – spontaneous release) X 100(%). All experiments were performed in triplicate.
2.14. Statistical Analysis
Figures were generated using GraphPad Prism 8.0 software (GraphPad Software, Inc., La Jolla, USA). Statistical analyses were also performed using this software. Differences between means were evaluated using the student’s t-test and were deemed significant at *p ≤ 0.05 **p ≤ 0.01.
4. Discussion
Current animal models of gastric cancer have certain limitations and cannot fully mimic the complexity of human gastric cancer [
45]. This has led us to use gastric cancer specimens obtained from patients to simulate in vivo conditions, reflecting the pathophysiology of human diseases more accurately. We found that lidocaine has potential applications in gastric cancer research, particularly due to its anti-tumor and anti-inflammatory effects. Given the limitations of gastric cancer animal models, using gastric cancer specimens from patients along with advanced culture techniques can provide a more accurate assessment of the efficacy and mechanisms of lidocaine.
This study investigates the role of lidocaine in modulating the immune response and its anti-tumor effect in gastric primary tumor-infiltrating immune cells (TIICs) within the tumor microenvironment (TME). The findings suggest that lidocaine regulates the suppressive activity of human Treg cells or alternative (M2) macrophages, providing new opportunities to improve the outcome of cancer immunotherapy. Given that long-term treatment effects of multiple immunotherapy approaches have been unsatisfactory in solid tumors, especially in gastric cancer, these findings hold promise for advancing treatment strategies [
46]. Gastric cancer may partly result from the immunosuppressive status in the TME mediated by negative immune cells such as Tregs or M2 macrophages, and immune inhibitory cytokines such as IL-10 and TGF-β [
46,
47]. However, little is known about cytokine expression in lidocaine-treated gastric TIICs. Thus, the present study investigates cytokine expression levels in lidocaine-treated gastric tumor-infiltrating immune cells (TIICs) to explore the potential role of lidocaine in anti-cancer activity within the gastric TME.
IL-10 is a well-known anti-inflammatory cytokine that limits the immune response during infections and is produced by nearly every type of cells in the immune system [
48]. It inhibits inflammatory cytokines [
49]. This study is the first to show that lidocaine causes an increase in IL-10 levels in lidocaine-treated CD4
+CD25
+ and CD14
+ PBMCs, while a decrease in IL-10 levels is observed in lidocaine-treated CD4
+CD25
+ and CD14
+ TIICs. CD14
+ tumor-infiltrating macrophages expressing IL-10 have been found and enriched in gastric cancer patients to facilitate immune evasion [
50]. Lidocaine suppresses IL-10 secretion in CD14
+ tumor-infiltrating macrophages and CD4
+CD25
+ tumor-infiltrating Tregs. Taken together, it is likely that IL-10 is involved in lidocaine-modulated mechanisms against gastric cancer. PD-1 have been shown to have inhibitory functions in T cells [
51]. Interleukin-10 receptor signaling promotes the maintenance of PD-1
+CD8
+ T cell population [
52]. The blockade of PD-1, combined with IL-10 neutralization, augmented the anti-tumor response [
53]. Our results demonstrate a decrease in PD-1 expression in lidocaine-treated CD8
+TIICs through NF-kb pathway, suggesting potential for therapy in gastric cancer.
Distinct from other members of the IL-12 family, IL-35 is a novel inhibitory cytokine that suppresses T cell proliferation [
52]. Recently, IL-35 has been proposed as a novel immunosuppressive cytokine [
54]. It has been demonstrated to promote tumor angiogenesis and inhibit the antitumor cytotoxic CD8
+ T cell response [
55]. In addition, IL-35 expression is considered to be associated with colorectal cancer progression and prognosis [
56]. We first found that lidocaine, in a dose-dependent manner, significantly reduces levels of IL-35 in CD4
+CD25
+ or CD14
+ TIICs. The present report analyzes the expression of IL-35 in lidocaine-treated PBMCs and TIICs. To the best of our knowledge, this is the first study to explore the function of the novel cytokine IL-35 in lidocaine-treated PBMCs and TIICs.
The present study suggests that in normal immune cells, lidocaine exerts its functions by inhibiting the production of IFN-γ and IL-12, while increasing the production of anti-inflammatory cytokines IL-10 and TGF-β, as well as a novel immunomodulator cytokine IL-35 from normal PBMCs. Our findings provide new insights into the anti-inflammatory mechanisms of lidocaine and a novel molecular target. In the TME, Tregs and tumor-infiltrating macrophages are considered to be sources of IL-35
[57]. It was reported that tumor-derived IL-35 increases tumorigenesis with a pro-tumor effect, and IL-35 production in the TME increases suppressor cells [
58]. Like TGF-β and IL-10, IL-35 can also induce the development of CD4
+CD25
+Foxp3
+ Treg population [
16]. CD4
+CD25
+Foxp3
+ Tregs are recruited and expanded in tumors and constitute an important mechanism utilized by tumor cells to evade protective immunity and support metastatic growth [
59]. Lidocaine exerts its functions on TME by inhibiting the production of IL-10 and TGF-β as well as a novel immunomodulator cytokine IL-35 from CD4
+CD25
+Foxp3
+TIICs. The differential anti-inflammatory and pro-inflammatory responses to lidocaine observed between TIICs and normal PBMCs may be attributed to the distinct microenvironments and functional states of these cell types. TIICs are directly exposed to the gastric tumor microenvironment, which can influence their response to lidocaine differently compared to PBMCs, which are typically isolated from normal peripheral blood and not directly exposed to tumor-associated factors.
Our findings reveal that lidocaine modulates the antitumor effects by decreasing IL-10, TGF-β, IL-35 level in CD4
+CD25
+Foxp3
+TIICs, decreasing PD-1 and increase IFN-g expression in cytotoxic CD8
+TIICs through NF-kb pathway. Lidocaine is a common local anesthetic; however, recent studies suggest that it induces apoptosis in gastric cancer cell lines in vitro [
44], although the use of 5 and 10 mM lidocaine has been found to be cytotoxic. We have found that lidocaine (0-1.5 mM) has no cytotoxicity in either TIICs or PGGCs. Lidocaine-untreated CD8
+TIICs showed minimal cell death of PGCCs, whereas lidocaine (1.5 mM)-treated CD8
+TIICs induced strong cell death. Lidocaine (1.5 mM)-treated CD8
+ TIICs displayed strong cell killing activity on PGCCs. The lidocaine (1.5 mM) blockade of PD-1 triggers cytotoxic CD8
+ T cell activation, resulting in the killing of PGCCs through immunogenic cell death. Administered local anesthetics such as lidocaine have favorable effects on overall gastric cancer patients’ TIICs. Lidocaine anesthesia may thus influence the TME of gastric cancer. This study further demonstrates that lidocaine may be involved in the anti-cancer effect of gastric cancer.
Figure 1.
Lidocaine reduces the secretion levels of IFN-γ by CD8+ PBMCs and IL-12 by CD14+ PBMCs. The effects of lidocaine (0.25 mM to 1.5 mM) on PMA and PHA-stimulated IFN-γ secretion from CD8+ PBMCs (A) were investigated. The IFN-γ level in the supernatant was determined at 72 hours by ELISA. Additionally, CD14+ PBMCs (B) were stimulated by PMA and PHA, then cultured in the absence or presence of graded concentrations of lidocaine. The IL-12 level in the supernatant was also determined at 72 hours by ELISA. Data are from distinct samples and are presented as the mean± SEM from three different experiments, each performed in duplicate. *P < 0.05, **P < 0.01. NC: negative control (non-stimulated cells).
Figure 1.
Lidocaine reduces the secretion levels of IFN-γ by CD8+ PBMCs and IL-12 by CD14+ PBMCs. The effects of lidocaine (0.25 mM to 1.5 mM) on PMA and PHA-stimulated IFN-γ secretion from CD8+ PBMCs (A) were investigated. The IFN-γ level in the supernatant was determined at 72 hours by ELISA. Additionally, CD14+ PBMCs (B) were stimulated by PMA and PHA, then cultured in the absence or presence of graded concentrations of lidocaine. The IL-12 level in the supernatant was also determined at 72 hours by ELISA. Data are from distinct samples and are presented as the mean± SEM from three different experiments, each performed in duplicate. *P < 0.05, **P < 0.01. NC: negative control (non-stimulated cells).
Figure 2.
Lidocaine induces the secretion of IL-10, TGF-β, and IL-35 by CD4+C25+ and CD14+ PBMCs. The effect of lidocaine on the secretion of IL-10 (A, B), TGF-β (C, D), and IL-35 (E, F) from CD4+CD25+ and CD14+ PBMCs was investigated. CD4+CD25+ and CD14+ PBMCs were cultured in the absence or presence of graded concentrations of lidocaine (0.25 mM to 1.5 mM). The levels of IL-10, TGF-β, and IL-35 in the supernatant were determined at 72 hours by ELISA. Data are from distinct samples and are presented as the mean± SEM from three different experiments, each performed in duplicate. *P < 0.05, **P < 0.01.
Figure 2.
Lidocaine induces the secretion of IL-10, TGF-β, and IL-35 by CD4+C25+ and CD14+ PBMCs. The effect of lidocaine on the secretion of IL-10 (A, B), TGF-β (C, D), and IL-35 (E, F) from CD4+CD25+ and CD14+ PBMCs was investigated. CD4+CD25+ and CD14+ PBMCs were cultured in the absence or presence of graded concentrations of lidocaine (0.25 mM to 1.5 mM). The levels of IL-10, TGF-β, and IL-35 in the supernatant were determined at 72 hours by ELISA. Data are from distinct samples and are presented as the mean± SEM from three different experiments, each performed in duplicate. *P < 0.05, **P < 0.01.
Figure 3.
The effect of lidocaine on IFN-γ secretion by CD8+ TIICs and IL-12 secretion by CD14+ TIICs. The effects of lidocaine on IFN-γ secretion from CD8+ TIICs (A) and IL-12 secretion from CD14+ TIICs (B) were investigated. CD8+ and CD14+ TIICs were stimulated in the absence or presence of graded concentrations of lidocaine (0.25 mM to 1.5 mM). The levels of IFN-γ and IL-12 in the supernatant were determined at 72 hours by ELISA. Results obtained from three different donors are shown. Data are from distinct samples and are presented as the mean± SEM. *P < 0.05, **P < 0.01.
Figure 3.
The effect of lidocaine on IFN-γ secretion by CD8+ TIICs and IL-12 secretion by CD14+ TIICs. The effects of lidocaine on IFN-γ secretion from CD8+ TIICs (A) and IL-12 secretion from CD14+ TIICs (B) were investigated. CD8+ and CD14+ TIICs were stimulated in the absence or presence of graded concentrations of lidocaine (0.25 mM to 1.5 mM). The levels of IFN-γ and IL-12 in the supernatant were determined at 72 hours by ELISA. Results obtained from three different donors are shown. Data are from distinct samples and are presented as the mean± SEM. *P < 0.05, **P < 0.01.
Figure 4.
The effect of lidocaine on IL-10, TGF-β, and IL-35 induced by CD4+CD25+ and CD14+ TIICs. The effect of lidocaine on IL-10 (A, B), TGF-β (C, D), and IL-35 (E, F) secretion from CD4+CD25+ and CD14+ TIICs were investigated. CD4+CD25+ and CD14+ TIICs were cultured in the absence or presence of graded concentrations of lidocaine (0.25 mM to 1.5 mM). The levels of IL-10, TGF-β, and IL-35 in the supernatant were determined at 72 hours by ELISA. Results obtained from three different donors are shown. Data are from distinct samples and are presented as the mean± SEM from three different experiments, each performed in duplicate. *P < 0.05, **P < 0.01.
Figure 4.
The effect of lidocaine on IL-10, TGF-β, and IL-35 induced by CD4+CD25+ and CD14+ TIICs. The effect of lidocaine on IL-10 (A, B), TGF-β (C, D), and IL-35 (E, F) secretion from CD4+CD25+ and CD14+ TIICs were investigated. CD4+CD25+ and CD14+ TIICs were cultured in the absence or presence of graded concentrations of lidocaine (0.25 mM to 1.5 mM). The levels of IL-10, TGF-β, and IL-35 in the supernatant were determined at 72 hours by ELISA. Results obtained from three different donors are shown. Data are from distinct samples and are presented as the mean± SEM from three different experiments, each performed in duplicate. *P < 0.05, **P < 0.01.
Figure 5.
Schematic diagram: lidocaine-modulated immunosuppression of normal PBMCs and anti-tumor effect of gastric TIICs. This diagram illustrates the dual effects of lidocaine on immune cells: 1. PBMCs: Demonstrates how lidocaine modulates immunosuppression in normal PBMCs, highlighting changes in cytokine secretion. 2. TIICs: Illustrates the anti-tumor effect of lidocaine on gastric TIICs, focusing on its impact on cytokine secretion.
Figure 5.
Schematic diagram: lidocaine-modulated immunosuppression of normal PBMCs and anti-tumor effect of gastric TIICs. This diagram illustrates the dual effects of lidocaine on immune cells: 1. PBMCs: Demonstrates how lidocaine modulates immunosuppression in normal PBMCs, highlighting changes in cytokine secretion. 2. TIICs: Illustrates the anti-tumor effect of lidocaine on gastric TIICs, focusing on its impact on cytokine secretion.
Figure 6.
Lidocaine inhibits IL-35, IL-10, and TGF-β production by CD4+CD25+Foxp3+ TIICs.Sorted CD4+CD25+TIICs were treated with 1.5 mM lidocaine for 72 hours followed by staining with anti-IL-35, anti-IL-10, anti-TGF-β, and anti-Foxp3 antibodies for flow cytometry analysis. Isotype controls were used to distinguish between positive and negative cells for IL-35, IL-10, TGF-β, and Foxp3. Typical flow cytometry dot plot analysis revealed the percentage of (A, B) CD35+Foxp3+CD4+CD25+ TIICs, (C, D) IL-10+Foxp3+CD4+CD25+ TIICs, and (E, F) TGF-β+Foxp3+CD4+CD25+ TIICs treated with lidocaine (1.5 mM). Data are from distinct samples and presented as the mean± SEM in three different experiments, each performed in duplicate. *P < 0.05, **P < 0.01.
Figure 6.
Lidocaine inhibits IL-35, IL-10, and TGF-β production by CD4+CD25+Foxp3+ TIICs.Sorted CD4+CD25+TIICs were treated with 1.5 mM lidocaine for 72 hours followed by staining with anti-IL-35, anti-IL-10, anti-TGF-β, and anti-Foxp3 antibodies for flow cytometry analysis. Isotype controls were used to distinguish between positive and negative cells for IL-35, IL-10, TGF-β, and Foxp3. Typical flow cytometry dot plot analysis revealed the percentage of (A, B) CD35+Foxp3+CD4+CD25+ TIICs, (C, D) IL-10+Foxp3+CD4+CD25+ TIICs, and (E, F) TGF-β+Foxp3+CD4+CD25+ TIICs treated with lidocaine (1.5 mM). Data are from distinct samples and presented as the mean± SEM in three different experiments, each performed in duplicate. *P < 0.05, **P < 0.01.
Figure 7.
Lidocaine enhances antitumor immunity by reducing PD-1 and increase IFN-γ expression on CD8+ TIICs through NF-κB signaling pathway. Gastric CD8+ TIICs treated with lidocaine (1.5 mM) were analyzed by flow cytometry. Single-cell suspensions obtained from sorted CD8+ TIICs were stained to detect IFN-γ (A, B) and PD-1 (C, D). Analysis of IFN-γ production and PD-1 expression by lidocaine treated-CD8+TIICs with NF-κB inhibitor. CD8+TIICs were incubated for 1 h with or without 10 μM BAY11-7082 and then treated with lidocaine for 72 h. IFN-γ and PD-1 was measured by flow cytometry. All flow cytometry analyses were gated on total live cells. Data are from distinct samples and presented as the mean± SEM. *p < 0.05, **p < 0.01; n ≥ 3.
Figure 7.
Lidocaine enhances antitumor immunity by reducing PD-1 and increase IFN-γ expression on CD8+ TIICs through NF-κB signaling pathway. Gastric CD8+ TIICs treated with lidocaine (1.5 mM) were analyzed by flow cytometry. Single-cell suspensions obtained from sorted CD8+ TIICs were stained to detect IFN-γ (A, B) and PD-1 (C, D). Analysis of IFN-γ production and PD-1 expression by lidocaine treated-CD8+TIICs with NF-κB inhibitor. CD8+TIICs were incubated for 1 h with or without 10 μM BAY11-7082 and then treated with lidocaine for 72 h. IFN-γ and PD-1 was measured by flow cytometry. All flow cytometry analyses were gated on total live cells. Data are from distinct samples and presented as the mean± SEM. *p < 0.05, **p < 0.01; n ≥ 3.
Figure 8.
Apoptotic effects of lidocaine on TIICs, PBMCs, PGCCs, and CD8+TIICs co-cultured with PGCCs assessed by flow cytometry. 5mM and 10mM lidocaine induce TIICs and PBMCs apoptosis but 0.5 and 1.5 lidocaine does not induce PGCCs apoptosis. TIICs (A), PBMCs (C) and PGCCs (E) were assessed by flow cytometric analysis using propidium iodide-stained cells. Firstly, 104 cells were incubated in 96-well plates in the presence or absence of the indicated concentrations of lidocaine. After 72 h treatment, cells were washed with PBS and fixed with 70% ethanol for 1 h on ice. Pelleted cells were incubated with RNaseA (0.1 mg/mL) and propidium iodide (40 μg/mL) for 1 h with shaking and protected from light. The percentage of subG1 population was determined by flow cytometry. (G) Flow cytometry assessment of cell death for lidocaine (1.5 mM)-treated CD8+TIICs, lidocaine (1.5 mM)-treated PGCCs or lidocaine (1.5 mM)-treated CD8+TIICs co-cultured with PGGCs. Data are representative of three independent experiments; n > = 3. (B)(D)(F)(H) Data are from distinct samples and presented as the mean± SEM in three different experiments, each performed in duplicate. *P < 0.05 **P < 0.01.
Figure 8.
Apoptotic effects of lidocaine on TIICs, PBMCs, PGCCs, and CD8+TIICs co-cultured with PGCCs assessed by flow cytometry. 5mM and 10mM lidocaine induce TIICs and PBMCs apoptosis but 0.5 and 1.5 lidocaine does not induce PGCCs apoptosis. TIICs (A), PBMCs (C) and PGCCs (E) were assessed by flow cytometric analysis using propidium iodide-stained cells. Firstly, 104 cells were incubated in 96-well plates in the presence or absence of the indicated concentrations of lidocaine. After 72 h treatment, cells were washed with PBS and fixed with 70% ethanol for 1 h on ice. Pelleted cells were incubated with RNaseA (0.1 mg/mL) and propidium iodide (40 μg/mL) for 1 h with shaking and protected from light. The percentage of subG1 population was determined by flow cytometry. (G) Flow cytometry assessment of cell death for lidocaine (1.5 mM)-treated CD8+TIICs, lidocaine (1.5 mM)-treated PGCCs or lidocaine (1.5 mM)-treated CD8+TIICs co-cultured with PGGCs. Data are representative of three independent experiments; n > = 3. (B)(D)(F)(H) Data are from distinct samples and presented as the mean± SEM in three different experiments, each performed in duplicate. *P < 0.05 **P < 0.01.
Figure 9.
Lidocaine-treated CD8+TIICs dependent immunogenic cell death of PGCCs.Lidocaine (1.5 mM)-treated CD8+TIICs, lidocaine (1.5 mM)-treated PGCCs or lidocaine (1.5 mM)-treated CD8+TIICs co-cultured with PGGCs at the 5:1 E:T ratios. Target cell cytotoxicity was determined at 2 hours by a DELFIA EuTDA assay. Data represent the mean of triplicate experiments, and experiments were repeated at least three times using different donor with similar results. Data are from distinct samples and presented as the mean± SEM. *p < 0.05, **p < 0.01 compared with mock treated cells; n > = 3.
Figure 9.
Lidocaine-treated CD8+TIICs dependent immunogenic cell death of PGCCs.Lidocaine (1.5 mM)-treated CD8+TIICs, lidocaine (1.5 mM)-treated PGCCs or lidocaine (1.5 mM)-treated CD8+TIICs co-cultured with PGGCs at the 5:1 E:T ratios. Target cell cytotoxicity was determined at 2 hours by a DELFIA EuTDA assay. Data represent the mean of triplicate experiments, and experiments were repeated at least three times using different donor with similar results. Data are from distinct samples and presented as the mean± SEM. *p < 0.05, **p < 0.01 compared with mock treated cells; n > = 3.
Figure 10.
Schematic diagram illustrating the lidocaine-mediated anti-tumoral mechanism through immunogenic cell death targeting PGCCs. Lidocaine inhibits the production of IL-35, IL-10, and TGF-β by CD4+CD25+Foxp3+ tumor-infiltrating immune cells (TIICs). Additionally, lidocaine enhances antitumor immunity by reducing PD-1 expression and increasing IFN-γ expression on CD8+ TIICs via the NF-κB signaling pathway. The lidocaine-treated CD8+ TIICs subsequently promote immunogenic cell death of PGCCs. Long→: Treated. Short→: Linked. ┬: Inhibition.
Figure 10.
Schematic diagram illustrating the lidocaine-mediated anti-tumoral mechanism through immunogenic cell death targeting PGCCs. Lidocaine inhibits the production of IL-35, IL-10, and TGF-β by CD4+CD25+Foxp3+ tumor-infiltrating immune cells (TIICs). Additionally, lidocaine enhances antitumor immunity by reducing PD-1 expression and increasing IFN-γ expression on CD8+ TIICs via the NF-κB signaling pathway. The lidocaine-treated CD8+ TIICs subsequently promote immunogenic cell death of PGCCs. Long→: Treated. Short→: Linked. ┬: Inhibition.