Back to Journals » Journal of Inflammation Research » Volume 18
The Role of Dectin-1-Akt-RNF146 Pathway in β-Glucan Induced Immune Trained State of Monocyte in Sepsis
Authors Guo C, Xu P, Luo W, Zhang J, Sun X , Hoang H, Ma D, Wu D, Zhong J , Miao C
Received 2 August 2024
Accepted for publication 12 January 2025
Published 24 January 2025 Volume 2025:18 Pages 1147—1165
DOI https://doi.org/10.2147/JIR.S482213
Checked for plagiarism Yes
Review by Single anonymous peer review
Peer reviewer comments 2
Editor who approved publication: Dr Adam Bachstetter
Chenyue Guo,1,* Peiyao Xu,1,* Wenchen Luo,1,* Jinlin Zhang,1,* Xingfeng Sun,2 Harry Hoang,3 Duan Ma,4 Dehua Wu,5 Jing Zhong,1 Changhong Miao1
1Department of Anesthesiology, Zhongshan Hospital Fudan University, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, People’s Republic of China; 2Department of Anesthesiology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, People’s Republic of China; 3Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland; 4Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, Collaborative Innovation Center of Genetics and Development, Institute of Biomedical Sciences, School of Basic Medical Sciences, Fudan University, Shanghai, People’s Republic of China; 5Department of Anesthesiology, Shanghai Songjiang District Central Hospital, Shanghai, People’s Republic of China
*These authors contributed equally to this work
Correspondence: Changhong Miao; Jing Zhong, Email [email protected]; [email protected]
Background: Sepsis is regarded as a dysregulated immune response to infections. Recent study showed partially reversal of immunosuppression by trained immunity, which fosters an enhanced immune response towards a secondary challenge. However, the role of trained immunity in sepsis has not been fully understood.
Methods: We profiled the characteristics of peripheral blood mononuclear cells from septic patients using single-cell RNA sequencing (scRNA-seq) analyses. Murine double-hit models (pretreatment or post-treatment of β-glucan in septic mice) and murine monocyte/macrophage cell line RAW264.7 were used then.
Results: scRNA-seq revealed that Ring finger protein 146 (RNF146) and protein kinase B (Akt) were downregulated in the immunosuppression period of septic patients and were verified to be decreased in bone marrow and spleen monocytes from septic mice. While β-glucan pretreatment improved the immunosuppressed state in septic mice and increased dectin-1/Akt/RNF146 expressions in monocytes, along with the increased survival rate, inflammatory factors and aerobic glycolysis, indicating a change from immunosuppression to immune training. Moreover, RNF146 regulated dectin-1-Akt-mTOR signaling in the trained immune state of murine monocyte/macrophage RAW264.7 cell line and the expression of RNF146 was dependent on dectin-1-Akt activation. The inhibition of dectin-1 by its antagonist laminarin downregulated Akt-RNF146 signaling and partially reversed β-glucan induced trained immunity in septic mice.
Conclusion: RNF146 and Akt are downregulated in the immunosuppression period of sepsis, while increased after β-glucan pretreatment induced trained immunity in septic mice. Moreover, RNF146 regulates the immune trained state of monocyte through dectin-1-Akt-mTOR pathway, suggesting a possible target in reversal of immunosuppression in sepsis.
Keywords: RNF146, β-glucan, trained immunity, immunosuppression, sepsis
Introduction
Sepsis is a life-threatening syndrome, characterized by multiple organ failure resulting from a dysregulated host response to infection.1–3 Despite rapid progresses made in the early diagnosis and treatment, the associated mortality of sepsis is still on the rise and can range anywhere from 30% to 50% depending on international registers.4 Sepsis is regarded as a dysregulated complex immune response to infections, inducing defects in both innate and adaptive immune and eventually leading to prolonged and significant immunosuppression.5–8 Specifically, blood leukocytes of septic patients have a diminished ability to release proinflammatory cytokines such as tumor necrosis factor (TNF), interleukin (IL)-1, IL-6, and IL-12 following stimulation ex vivo.9 Immunosuppression represents one extreme in the spectrum of innate immune memory and can be induced by high bacterial burden in vivo or lipopolysaccharide (LPS) exposure in vitro9 and controlled by downregulated protein kinase B (Akt)-hypoxia-inducible factor 1 alpha (HIF-1α) pathway.10 Immunosuppression leads to attenuated immune responses to secondary stimulus, so it is possible to target the immune system to promote host fitness and survival in patients with sepsis.11
Reversal of immunosuppression using innate immune “trainers” is a viable therapeutic strategy.12 Trained immunity, defined by the boosted innate immune response after experiencing an initial stimulus, was first proposed by professor Netea in 2011 and is a powerful defense against infection, regardless of bacteria, fungi or virus.9 Trained immunity can be induced by β-glucan or oxidized low-density lipoprotein (oxLDL), resulting in epigenetic reprogramming of myeloid cells, especially in monocytes and macrophages.9 Boris et al reported that β-glucan was capable of reversing macrophage tolerance induced by ex vivo LPS exposure,12 leading to a reversal inflammatory status13 and antimicrobial activity.14 As trained immunity can deliver nonspecific protection from re-infection, it may provide a new direction for preventing and treating immunosuppression in sepsis.15 Therefore, inducers of trained immunity are of great interest as a treatment approach to reverse immunosuppression states in sepsis.
In this study, we aimed to investigate the immune suppressed state of sepsis and the role of trained immunity in sepsis-induced immunosuppression and the underlying mechanism. scRNA-seq analysis was conducted in septic patients, and then the potential role of trained immunity induced by β-glucan in sepsis-induced immunosuppression and the involvement of relevant pathways were investigated in murine models and murine monocyte/macrophage RAW264.7 cell lines.
Material and Methods
Patients
Our study was approved by the ethics committee of Zhongshan Hospital Fudan University (B2021-215R) and was registered at the Chinese Clinical Trial Registry (ChiCTR2100048148). Our study was complied with the Declaration of Helsinki. Patients who experienced sepsis induced by Gram-negative bacterial infection were recruited from the Intensive Care Unit (ICU) of Zhongshan Hospital, Fudan University, and all subjects were included from July to December 2021. Informed consent was obtained from the participants, or the person authorized by the participants. Patients with sepsis were required to fulfill the diagnostic criteria for sepsis-3.16 Patients with immunosuppression were required to fulfil the following criteria: C-reactive protein (CRP) > 150 μg/dL; total lymphocyte count (LYM) < 0.8×109/L; serum albumin < 30 g/L; prealbumin level < 0.1 g/L; retinol-binding protein (RBP) < 1 mg/dL; creatinine height index (CHI) < 80%; weight loss > 10% or Body Mass Index < 18 during hospitalization. Exclusion criteria included: 1) immunodeficient persons and 2) those receiving immunomodulatory medications (including prednisone or steroid analogs, chemotherapy, or biological immunomodulators).
Single-Cell Preparation and Single-Cell RNA Statistical Analysis
Peripheral blood mononuclear cells (PBMCs) were isolated from whole-blood samples by density-gradient centrifugation, as described previously.17 Cells from each sample were labeled with sample tags (TotalSeqTM–C antibodies), following the manufacturer’s protocol. The scRNA-seq and V(D)J libraries were generated using the 10× Genomics Chromium Controller Instrument and Chromium Single Cell 5′ library and gel bead kit, along with the V(D)J enrichment kit (10× Genomics, Pleasanton, CA). The scRNA-seq data analysis was performed by NovelBio Co., Ltd. with the NovelBrain Cloud Analysis Platform (www.novelbrain.com). In brief, we applied fastp18 with the default parameters to filter the adaptor sequences and remove low-quality reads to achieve clean data. Then, feature-barcode matrices were obtained by aligning reads to the human genome (GRCh38 Ensemble: version 91) using CellRanger v3.1.0. The Seurat package (version: 3.1.4, https://satijalab.org/seurat/) was used for batch correction based on the expression table according to the UMI counts of each sample and percentage of mitochondria rate to obtain scaled data. Using the graph-based cluster method, we calculated marker genes using the FindAllMarkers function with the Wilcox rank sum test algorithm under the following criteria: lnFC > 0.25; P-value < 0.05; min.pct > 0.1. All single-cell RNA sequencing data were deposited in the Gene Expression Omnibus (GEO: GSE217906).
Animals
Ten-week-old male C57BL/6 mice were purchased from animal experiment center of Fudan University, and maintained under pathogen-free conditions. All procedures were conducted in accordance with the guidelines of the Chinese council on animal care and we obtained approval from the animal review committee of Fudan University Shanghai Cancer Center (FUSCC-IACUC-S2023-0141). All surgeries were performed under anesthesia, and all efforts were made to minimize animal suffering.
Double-Hit Models
Two types of double-hit mouse models (pretreatment or post-treatment models) were used. Pretreatment double-hit model was constructed with β-1,3 (D) glucan (BG) (Invivo Gen, San Diego, USA) or 0.9% sodium chloride (NS) administration 7 days before cecal ligation and puncture (CLP) in mice. Specifically, mice received 2 mg/kg β-glucan or equal volume of NS intraperitoneally, and then CLP procedure was performed 7 days later (BG+CLP group, NS+CLP group). While post-treatment double-hit model was constructed with CLP procedure first, and 14 days later, 2mg/kg β-glucan or equal volume of NS was injected intraperitoneally (CLP+BG group, CLP+NS group), which is in line with previous study.19 The Mouse Sepsis Score (MSS) and weight were measured each day. MSS assessed the severity of fecal-induced peritonitis concluding seven variables and 0–28 points from mild to severe.
Monocyte Harvesting
C57BL/6 mice were euthanized via cervical dislocation and spleen monocytes and bone marrow mononuclear cells (BMMC) were harvested by following manners. The spleen was dissected and weighed. For dissociation of splenic tissue, the spleen was passed through a 70 µm cell strainer. Mouse splenocytes were centrifuged and resuspended in red blood cell lysis buffer (ACK lysis buffer) (Servicebio, Wuhan, China) for red blood cell lysis. Bone marrow was extracted from femura and tibiae of mice as follows. Bones were prepared by removing all soft tissue without fracturing, followed by a decapping of the bone on both sides. Subsequently, bone marrow was flushed out with pre-warmed DMEM (HyClone, Thermo, Waltham, MA, USA) media. Then extracted mouse splenocytes and myeloid cells were cultured at 37°C in a 5% CO2 incubator and were maintained in DMEM supplemented with 10% FBS (HyClone, Thermo, Waltham, MA, USA) and penicillin and streptomycin (P/S) (Gibco, Thermo, Waltham, MA, USA). Monocyte isolation was carried out by Moflo XDP Cell Sorter (Beckman Coulter, California, USA) according to the instructions provided by the manufacturer. Mouse splenocytes and myeloid cells extracted from mice were suspended respectively in FACS buffer (Phosphate Buffered Saline (PBS) with 1% BSA and 0.1% azide), and 1 µL/sample of Fc Block (BD Pharmingen, San Jose, CA, USA) was added to prevent nonspecific binding. The above suspended monocyte cell types were isolated using the following antibody combinations: PE anti-mouse Ly-6G (BioLegend, 127608, San Diego, CA, USA) and pacific blue anti-mouse CD11b (BioLegend, 101224, San Diego, CA, USA). The isolated mouse splenocytes and BMMC were then cultured in DMEM supplemented with 10% FBS and P/S at 37°C.
Flow Cytometry
Flow cytometry was used to determine the levels of intermediate monocytes (Ly6CintCX3CR1hi) from the BMMC and splenocytes isolated from septic or control mice, in order to identify the inflammatory monocytes, which were also identified as monocyte subsets with trained immunity phenotype.20,21 Isolated mouse splenocytes and BMMC were stained with PE anti-mouse CX3CR1 (BioLegend, 149005) and FITC anti-mouse Ly-6C (BioLegend, 128005) at 4°C for 30 min in the dark. Stained cells were analyzed by Cytomics FC 500 (Beckman Coulter, California, USA). Data were analyzed using FlowJo version 10.1 (Ashland, OR, USA).
Cytokine Measurements
Cytokine productions were assessed in supernatants of BMMC and spleen monocytes collected from septic or control mice using commercial Enzyme-Linked Immunosorbent Assay (ELISA) kits for mouse IL-10 (R&D systems, MN, USA), IL-6 (R&D systems, MN, USA), IL-1β (R&D systems, MN, USA) and TNF-α (R&D systems, MN, USA), in accordance with the manufacturer’s instructions.
Mitochondrial Respiration and Glycolysis Analysis
To analyze mitochondrial respiration and glycolysis, oxygen consumption rate (OCR, a measurement of oxidative phosphorylation) and extracellular acidification rate (ECAR, a measurement of glycolysis) were detected with Seahorse XF analyzer (Agilent Technologies, California, USA) in accordance with the manufacturer’s instructions. Spare respiratory capacity = OCR between carbonyl cyanide-4-(trifluoromethoxy)-phenylhydrazone (FCCP) and rotenone/antimycin A −OCR before oligomycin. Glycolytic reserve = ECAR between oligomycin and 2-deoxy-glucose (2-DG) −ECAR between glucose and oligomycin.
Cell Viability Assay
Isolated mouse splenocytes from septic or control mice were seeded in 96-well plate at densities of 5000 cells per well. A cell viability assay was conducted using the methyl tetrazolium (MTT) cell proliferation assay kit (R&D systems, MN, USA). 0.5 mg/mL of MTT was added to each well, and the plate was incubated at 37°C for 4 h. Then, the supernatant-containing MTT was removed, and 200 μL of dimethyl sulfoxide (DMSO) was added to each well. The plate was shaken thoroughly for 1 min on a shaker. The absorbance of the samples was measured at 570 nm by using a microplate reader (Bio-TEK Inc, Winooski, VT).
RNA Extraction, Quantitative Real-Time PCR (qPCR) and Western Blot Analysis
Total RNA was extracted using TRIzol (Invitrogen, Grand Island, NY, USA) and used to synthesize complementary DNA (cDNA) with PrimeScript RT reagent (TaKaRa) according to the manufacturer’s instructions. qPCR was then performed using Hieff UNICON® qPCR TaqMan Probe Master Mix (Yeasen, Shanghai, China) in an ABI7500 Real-Time PCR system (Applied Biosystems). The primers used in the present study are shown in Supplementary Table 1. Protocol for Western blot analysis was shown in Supplementary Materials.
Reagents and Plasmids
Lipofectamine 2000 were purchased from Invitrogen (Carlsbad, CA, USA). Anti-Ring finger protein 146 (RNF146) was purchased from Novus Biologicals (CO, USA). Anti-c-type lectin domain family 7 member A (dectin-1) mAbs was purchased from R&D systems (MN, USA). Anti-mammalian target of rapamycin (mTOR), anti-phosphorylated-mammalian target of rapamycin (p-mTOR), anti-Akt, anti-phosphorylated Akt (p-Akt) and anti-glyceraldehyde-3-phosphate dehydrogenase (GAPDH) mAbs were purchased from Proteintech (Chicago, IL, USA). Plasmid expressing pCDNA3.1-RNF146, siRNA RNF146, pCDNA3.1-dectin-1, and siRNA-dectin-1 (shown in Supplementary Table 2) was purchased from Genechem (Shanghai, China). We purchased plasmids expressing the constitutively active form of Akt (#10841, Addgene) and dominant negative form of Akt (#16243, Addgene) (shown in Supplementary Table 2).
Training State Model of RAW264.7 Cells
RAW264.7 cells (SIBS, Shanghai, China) were grown in DMEM with 10% FBS and penicillin and streptomycin and were incubated at 37°C with 5% CO2. RAW264.7 cells in the logarithmic growth stage were collected for subculture and further experimentation. RAW264.7 cells (P3, 1.71*106/mL) were treated with 100 ng/mL β-glucan for 24 h, and then washout and rested for 3 days in DMEM with 10% FBS followed by 100 ng/mL LPS stimulation for 24 h. The expressions of IL-1β, IL-6, TNF-α and IL-10 secreted by RAW264.7 cell were determined by ELISA (R&D systems, MN, USA) to confirm the training state.22
Immunofluorescence
RAW264.7 Cells were treated with 100 ng/mL β-glucan for 24 h, and then fixed with 4% paraformaldehyde in PBS, 2% bovine albumin (BSA) (Sigma-Aldrich), and 0.1% Triton X-100 (Sigma-Aldrich) for 1 h at room temperature. Then incubated with combinations of primary antibodies against RNF146, Akt (depending on the experiment) at 4°C overnight. Then, the fixed samples were washed with PBS containing 0.1% Triton X-100 and incubated with corresponding secondary antibodies conjugated with fluorescent dye at room temperature for 1 h. Fluorescent images were obtained using a confocal microscope (LSM 710, Zeiss).
Co-Immunoprecipitation (Co-IP)
The lysates of RAW264.7 were extracted via immunoprecipitation (IP) lysis buffer, cultivated with the indicated antibodies and negative control IgG all night at 4°C, followed by incubation with secondary antibody Horseradish Peroxidase (HRP) Mouse Anti-Rabbit IgG light chain (Abbkine, A25022). After mixing with beads, the complex was rinsed in IP buffer for 3 times and boiled with SDS buffer, followed by elution and Western blotting.
Administration of Dectin-1 Receptor Antagonist in vivo
The dectin-1 receptor antagonist laminarin (LAM) (Sigma-Aldrich, L9634) was diluted to 10 mg/mL in vehicle (saline) according to the manufacturer’s protocol. Then, LAM at 300 mg/kg/day or the same volume of NS was intraperitoneally injected once daily on the 7th day after CLP for the 2 subsequent days in double-hit mice model.
Statistical Analysis
We applied at least five samples for each experiment. Statistical evaluation was conducted using the GraphPad Prism software, version 9.5 (GraphPad, San Diego, CA, USA). Data were expressed as mean ± standard deviation (SD). For two-group comparisons, the unpaired two-tailed Student’s t test was used. One-way analysis of variance (or Kruskal–Wallis for non-parametric data) was used to assess significant differences among more than two groups (ANOVA; more than two groups), followed by Bonferroni’s multiple comparisons. Differences between survival curves were analyzed by the Log-Rank Mantel-Cox test. Differences of MSS and weight were analyzed by the Two-way ANOVA repeated measure, followed by Sidak’s or Dunnett multiple comparisons. P<0.05 was considered significant.
Results
scRNA-Seq Revealed Changes in Monocyte from Septic Patients
We profiled the characteristics of PBMCs using scRNA-seq analyses from the patients in the immunosuppression period of sepsis (n=4), acute period of sepsis (n=2), and healthy individuals (n=3) (shown in Supplementary Table 3). The cell compositions of the groups were identified and immune signatures were profiled using scRNA-seq on the 10×Genomics 5′ sequencing platform (shown in Figure 1a). A total of 23 cell clusters were identified (shown in Figure 1b). The percentages of monocytes in the immunosuppression period of sepsis, acute period of sepsis, and healthy control groups were 11.7%, 18.2%, and 11.8% (shown in Figure 1c) separately. We specifically re-clustered monocytes and regrouped them into four distinct clusters including classical monocytes (CD14 and VCAN), monocyte-platelet aggregates (MPA) (PPBP and PF4), IL-1β+ monocytes (IL-1B), and macrophages (FCGR3A) (shown in Figure 1d). The expression of marker genes of each cluster is also shown in Figure 1d. The percentages of the four clusters varied significantly in the patients undergoing the immunosuppression period of sepsis (shown in Figure 1e), during which, the classical monocyte, which is the main cell subset of monocytes, decreased significantly. The percentages of classical monocytes in the immunosuppression period of sepsis, acute period of sepsis, and healthy control groups were 68.82%, 79.73%, and 79.02% (shown in Figure 1e), respectively. The proportions of MPA had a big increase only in the immunosuppression period of sepsis. These cells expressed CD14 (marker of monocytes), PPBP and PF (markers of platelets), and played crucial roles in the signal pathways of platelets degranulation, activation and coagulation (shown in Supplementary Figure 1).
Expressions of genes related to immune functional features were differentially affected in immunosuppressed and proinflammatory septic patients compared to healthy controls. Notably, RNF146, a novel Poly (ADP) ribosylation-directed ring finger E3 ubiquitin ligase and Akt were downregulated in the immunosuppression period of the sepsis group compared with acute sepsis group and healthy individuals. Akt plays an important role in macrophage-mediated innate immunity and is involved in regulating immune response during sepsis. Previous studies reported that activation of the Akt signaling pathway promotes RNF146 expression in Parkinson’s disease23 and activated RNF146 enhances the glycolysis via the Akt signaling Pathway.20 To determine the changes in expressions of RNF146 and Akt in the immunosuppression period of sepsis, we next detected the relative genes expressions in BMMC and spleen monocytes obtained from the CLP-induced immunosuppressed mouse.
Akt and RNF146 Expressions Were Down-Regulated in Bone Marrow and Spleen Monocytes from Septic Mice
First, we determined the CLP-induced immunosuppressed mouse model. We found that monocytes from CLP mouse model yielded reduced IL-1β, IL-6, TNF-α and increased IL-10 levels 14 and 21 days after CLP (shown in Supplementary Figure 2a and b). The CLP-induced immunosuppression in the mouse model was verified by testing the responsiveness of BMMC and spleen monocytes to LPS stimulation in vitro (shown in Figure 2a) and splenocyte apoptosis.21 We found that BMMC exhibited reduced responsiveness to LPS stimulation in vitro, including decreased expression of pro-inflammatory cytokines (shown in Figure 2b) and decreased OCR and ECAR, indicating decreased spare respiratory capacity and decreased glycolytic reserve (P<0.001 and P<0.001 separately, shown in Figure 2c). Cytokine secretion and metabolism changes in spleen monocytes were found to be similar to those in BMMC (shown in Figure 2d and e). Moreover, CLP caused significant splenocyte cell viability impairment compared with the control (P<0.001, shown in Figure 2f), indicating CLP induces splenocyte apoptosis. The above changes suggested the immune suppressed state of monocytes in septic mice.
We next detected the mRNA levels of RNF146 and Akt pathway according to the results of scRNA-seq. Akt-mTOR pathway has been shown to regulate immune response during sepsis.24 Moreover, the activation of the Akt-mTOR pathway is essential for the maintenance of immune homeostasis in sepsis.15 Signal transducer and activator of transcription 3 (STAT3) regulates neutrophil release to help contain infection and limit inflammatory responses.25 Hence, the mRNA levels of RNF146, Akt, mTOR, and STAT3 in BMMC from CLP-induced immune suppressed septic mice and control mice were detected. We found that RNF146, Akt and mTOR mRNA levels were significantly decreased in BMMC from immunosuppressed septic mice compared with control ones (P<0.001 for all comparisons, shown in Figure 2g). However, the mRNA level of STAT3 was not significantly different between the two groups (P=0.736, shown in Figure 2g). We then detected the expressions of RNF146, p-Akt, and p-mTOR in BMMC from CLP-induced immunosupressed septic mice using Western blot and found that the expressions of RNF146, p-Akt and p-mTOR were significantly downregulated (P=0.003, P<0.001 and P=0.001 separately, shown in Figure 2h). These results identified the down-regulation of RNF146 and Akt pathway in the immunosuppressed state of septic mice.
β-Glucan Pretreatment Improved the Immune Suppressed State in Septic Mice and Increased Dectin-1/Akt/RNF146 Expressions
Immunosuppression in sepsis was reported to partially reversed by β-glucan induced trained immunity.12 To demonstrate the effect of trained immunity on the immune suppressed state in sepsis, we first developed a β-glucan induced trained immunity mouse model in accordance with previous research26 (shown in Supplementary Figure 3a). We found increased levels of intermediate monocytes (Ly6CintCX3CR1hi), which is the trained immunity phenotype of monocyte,27,28 increased levels of IL-1β, IL-6 and TNF-α, decreased IL-10, decreased OCR, and increased ECAR in mouse BMMCs and spleen monocytes, indicating the immune trained state of mouse model (shown in Supplementary Figure 3b–3f). Next, we used two types of double-hit mouse models to test the effect of trained immunity on immunosuppression in septic mice. We established the double-hit model in septic mice with β-glucan treatment before or after CLP (shown in Figure 3a). The β-glucan pretreatment group (BG+CLP group) showed a significantly higher survival rate than the control group (NS+CLP group) (P=0.018, shown in Figure 3b); while no difference was found in the β-glucan post-treatment group (CLP+BG group) and control group (P=0.542, shown in Figure 3c). Additionally, MSS, indicative of the severity in sepsis, was much lower in BG+CLP group than that in NS+CLP group (P<0.05 from the 10th day, shown in Figure 3b), and no difference was found in the CLP+BG group and CLP+NS group until the 27th day (P=0.023, shown in Figure 3c). Loss of body weight, attributed to ongoing catabolism in septic mice, was less in BG+CLP group than that in NS+CLP group (P<0.05 from the 14th day, shown in Figure 3b); while no difference was found in the CLP+BG group and CLP+NS group until the 25th day (P=0.024, shown in Figure 3c). Furthermore, spleen was found to be both larger and heavier in BG+CLP group than that in NS+CLP group (P<0.001, shown in Supplementary Figure 4a). Collectively, our data indicated that β-glucan pretreatment improves the survival rate of septic mice, and reduces the severity of sepsis. β-glucan pretreatment also resulted in enhanced inflammatory cytokines secretion, with increased levels of IL-1β, IL-6 and TNF-α release (P<0.001, P<0.001 and P=0.003 in BMMC supernatant; P<0.001, P<0.001 and P=0.003 in spleen monocytes supernatant, respectively), and deceased IL-10 release (P<0.001 in BMMC supernatant; P<0.001 in spleen monocytes supernatant, shown in Figure 3d and Supplementary Figure 4b). Furthermore, β-glucan pretreatment induced increased ECAR (increased glycolytic reserve in BMMC and spleen monocytes, P<0.001 and P=0.019 separately) and decreased OCR (decreased spare respiratory capacity in BMMC and spleen monocytes, P=0.001 and P<0.001 separately) in monocytes (shown in Figure 3e and Supplementary Figure 4c), indicating the enhanced glycolysis which is the characteristic metabolic change of trained immunity.
We then detected the expressions of RNF146 and Akt pathway in monocytes from β-glucan preteated septic mice. The dectin-1 was described as the specific β-glucan receptor expressed on myeloid cells, with crucial functions in immune responses including pro-inflammatory responses such as cytokine production, reactive oxygen species generation and phagocytosis.11 The induction of aerobic glycolysis through dectin-1-Akt pathway represents the metabolic cornerstone of trained immunity.15 The activation of dectin-1 by β-glucan can induce the expression of p-Akt,14,29 and trigger downstream Akt signaling pathway.15 Therefore, we next examined the expressions of dectin-1, p-Akt and RNF146 in BMMC and spleen monocytes from β-glucan pretreated septic mice. Immunoblotting analysis revealed that the expressions of RNF146, p-mTOR, p-Akt and dectin-1 were upregulated in monocytes from septic mice with β-glucan pretreatment, which were also recognized as immune trained state in mice (shown in Figure 3f and g).
RNF146 Regulated Dectin-1-Akt-mTOR Signaling in the Trained Immune State of Murine Monocyte/Macrophage RAW264.7 Cell Line
RNF146 has been reported to be a key regulator in cell survival, energy metabolism, and immune function during both physical and pathological processes.30,31 To further investigate the role of RNF146 in β-glucan induced trained immunity, murine monocyte/macrophage cell-line RAW264.7 was used. The immune-trained state of RAW264.7 was induced by β-glucan treatment for 24 h, and 3 days later, cells were treated with LPS for an additional 24 h (shown in Figure 4a). siRNA RNF146 was used to downregulate the expression of RNF146, and we found the decreased secretion of IL-1β, IL-6, TNF-α (P=0.002, P=0.002 and P<0.001, respectively) and increased secretion of IL-10 (P=0.048) in β-glucan pretreated RAW264.7 cells (shown in Figure 4b), while overexpression of RNF146 led to the opposite (P=0.020, P=0.037, P<0.001, and P<0.001 respectively) (shown in Figure 4c). Then OCR and ECAR analysis was performed and findings showed that knockdown of RNF146 resulted in decreased aerobic glycolysis activation (increased in spare respiratory capacity of OCR; decreased in glycolytic reserve of ECAR; P=0.026 and P=0.011, respectively, shown in Figure 4d and e), while the overexpression of RNF146 led to the opposite (decreased spare respiratory capacity of OCR; increased glycolytic reserve of ECAR, P<0.001 and P=0.012, respectively, shown in Figure 4d and e). Collectively, these results indicate that RNF146 regulates the trained immune state induced by β-glucan in monocyte/macrophage.
To further investigate the pathway, we transfected RAW264.7 with siRNA RNF146, which was treated with β-glucan and LPS, and found that knockdown of RNF146 restrained the activation of p-mTOR (shown in Figure 5a and b), while RNF146 overexpression enhanced the activation of p-mTOR (shown in Figure 5c and d). Interestingly, the expressions of p-Akt and dectin-1 increased after β-glucan and LPS treatment and were not affected by RNF146 (shown in Figure 5a–d). Furthermore, knockdown of dectin-1 using siRNA dectin-1 resulted in lower expressions of p-mTOR, p-Akt and RNF146 in β-glucan and LPS-treated RAW264.7, while dectin-1 overexpression enhanced the expressions of p-mTOR, p-Akt and RNF146 (shown in Figure 5e). We then knocked down the dectin-1 combined with the overexpression of RNF146 in RAW264.7, and found that p-mTOR increased after β-glucan and LPS treatment and the expression of p-Akt decreased, while the overexpression of dectin-1 and knockdown of RNF146 led to the opposite (shown in Figure 5f). Taken together, these results demonstrated that RNF146 might regulate the trained immune state of monocyte/macrophage via dectin-1-Akt-mTOR pathway.
Since RNF146 may serve as a regulator in the Akt pathway in trained immune state of RAW264.7, we then examined whether Akt activation alone is sufficient for RNF146 expression. To induce Akt activation, we generated a constitutively active form of Akt (Akt-CA) in RAW264.7 cells, with a dominant negative form of Akt (Akt-DN) serving as a negative control. The results showed that the forced activation of Akt through Akt-CA expression is sufficient to induce RNF146 expression (P<0.001) (shown in Figure 6a and b). Additionally, immunofluorescence analysis and co-immunoprecipitation (Co-IP) assay showed that Akt and RNF146 activated by β-glucan can be co-localized in RAW246.7 (P<0.001 and P<0.001, separately) (shown in Figure 6c and d), and RNF146 was immunoprecipitated (IP) by Akt (shown in Figure 6e), indicating an interaction between RNF146 and Akt. Together, these results strongly suggest that RNF146 serves as a downstream of decin-1 and Akt. RNF146 may function as a substrate of Akt and play a role in modulating trained immunity induced by β-glucan, depending on Akt activation.
Blockade of Dectin-1 Diminished Trained Immunity Induced by β-Glucan and Inhibited Akt-RNF146 Signaling in Mice
In order to further investigate the potential role of dectin-1-Akt-RNF146 pathway in trained immunity, the dectin-1 antagonist LAM was used in mice (shown in Figure 7a). After LAM treatment, the survival rate of β-glucan pretreatment mice (BG+CLP+LAM group) was not higher than the control (NS+CLP+NS group and NS+CLP+LAM group) any more (P=0.634 and P=0.463 separately) (shown in Figure 7b). And there was also no difference in MSS after 3 days of LAM injection (P=0.256 on the 18th day) and in weight after 2 days of LAM injection (P=0.071 on the 17th day) between the BG+CLP+LAM group and the NS+CLP+NS group (shown in Figure 7b). But the MSS after CLP in the NS+CLP+LAM group was significantly higher than that in the BG+CLP+LAM group except on the 18th day (P=0.092 on the 18th day, P<0.05 for other comparisons) (shown in Figure 7b). Besides, after the LAM injection, the MSS was much higher and the loss of weight was more in BG+CLP+LAM group than BG+CLP+NS group (P<0.01 for all comparisons) (shown in Figure 7b). In comparison to the BG+CLP+NS group, decreased levels of intermediate monocytes (Ly6CintCX3CR1hi) were observed from the BG+CLP+LAM group, indicating that blockade of dectin-1 reduces the inflammatory phenotype of monocyte (P<0.05) (shown in Figure 7c). LAM treatment significantly decreased IL-1β, IL-6, TNF-α release and increased IL-10 release from BMMC and decreased p-mTOR, p-Akt and RNF146 expression in BMMC from mice in BG+CLP+LAM group compared with BG+CLP+NS group (P<0.001 for all comparisons) (shown in Figure 7d–f). Taken together, these results indicated that inhibition of dectin-1 diminishes the trained immunity induced by β-glucan in septic mice and the expressions of p-Akt and RNF146 in BMMC are decreased, providing evidence for the potential involvement of dectin-1-Akt-RNF146 signaling in trained immunity.
Discussion
In this study, our scRNA-seq results showed that RNF146 and Akt were downregulated in the immunosuppression period of septic patients, while β-glucan-induced trained immunity increased the expressions of RNF146 and Akt in monocytes from immune suppressed septic mice. We discovered a previously unknown role of RNF146 in regulating immune trained state of monocyte/macrophage via dectin-1-Akt-mTOR pathway. Moreover, β-glucan-induced trained immunity reduced the severity of sepsis and improved the survival rate of septic mice.
In sepsis, CLP-induced immunosuppression of innate immune cells is known to contribute to immune paralysis and metabolic reprogramming,7 which places the individual at greater risk of opportunistic infections.32,33 Previous animal studies approached the concept of “post-septic immunosuppression” using double-hit models, and those studies analyzed the susceptibility to a second stimulus from days to months after the insult.34–37 In this study, we demonstrated CLP can be done after 7 days pretreatment with β-glucan to construct a double-hit mouse model, which was in accordance with previous research.37
Sepsis has long been recognized for suppressive influence on hematopoiesis, resulting in myelosuppression.33,38, In the past decade, emerging evidence has shown long-term adaptation9,35 of the innate immune system through epigenetic and metabolic programming of myeloid cells,36 resulting in hyper-responsiveness upon re-stimulation in these cells. Meanwhile, there is growing evidence that sepsis induces a trained immunity phenotype in BMMCs after the initial insult via functional reprogramming, which refines the current hypothesis of a persisting global post-septic immunosuppression on the cellular level.39,40 In this study, we found an enhanced inflammatory response and increased aerobic glycolysis in isolated bone marrow and spleen monocytes from β-glucan pretreated, but not post-treated septic mice, which was in accordance with the characteristics of immune-trained monocytes. β-glucan induced trained immunity is similar to the clinical Bacillus Calmette Guerin (BCG) vaccine injection. Our study suggested that β-glucan pretreatment can improve the immunosuppressed state of sepsis and reprogram the metabolism of monocytes. The suppression period of sepsis is usually in the late period of sepsis and may last for a period of time depending on the patient’s condition. Therefore, β-glucan-like treatment may be effective for septic patients with immunosuppression and can be applied in high-risk patients. The effectiveness of β-glucan as immunoadjuvants in sepsis was observed in the patients with Sars-Cov-2 infection.41 While there are different chemical compositions of β-glucan and the processing for extraction and purification varies based on their functional properties, the biological response-modifying actions of its effect on septic immunosuppression need further study.42,43 Thus, it is necessary to explore the effect of β-glucan-induced trained immunity on immunosuppression during sepsis and the potential mechanisms.
Induction of trained immunity is mediated by activation of immune and metabolic pathways.44 The glycolytic phenotype is linked to the immune response.45 Metabolism is a guiding force for trained immunity.46 Aerobic glycolysis in the trained monocytes might induce pro-inflammatory cytokine production.15 For example, glycolysis is necessary for IL-1β expression in inflammatory diseases and promotes inflammatory activation. Activated glycolytic phenotype HIF-1α regulates inflammatory gene IL-1β induction.15,45,47 Recent research also highlights the physiological role of cytokines in the immune-metabolic interplay. IL-1β was reported to be activated to allocate glucose to immune cells, and TNF is activated from lipolysis and induces insulin resistance.47 However, the hydrolysis of glycogen was also reported to produce several products which inhibited or reduced production and secretion of proinflammatory cytokines such as IL-1β, IL-6 and TNF-α in inflammatory diseases.48 Therefore, when discussing the immune training effect of β-glucan, the observed changes in cytokines in this study are due to glycolysis or immune training itself, which deserves further study.
Changes in the inflammatory capacity of monocytes are associated with altered metabolic processes.7 A key part of metabolic rewiring of innate immune cells is the switch from oxidative phosphorylation to aerobic glycolysis.49,50 The axis involving dectin-1 (known as a β-glucan receptor and widely expressed on the surface of the myeloid lineage51), Akt and mTOR are involved in this metabolic shift,15 and play a crucial role in reducing basal respiration rate, increasing glucose consumption, and higher lactate production. Transcriptional upregulation of genes involved in glycolysis as well as genes involved in mTOR signaling, a key regulator of glucose metabolism, are responsible for the enhanced glycolytic activity.15,52 In our study, β-glucan pretreatment induced aerobic glycolysis in BMMC and led to the increased expression of p-mTOR, p-Akt and RNF146, indicating the involvement of Akt/mTOR pathway and RNF146 in the process. Our scRNA-seq results also suggested the potential role of RNF146 and Akt pathway in the immunosuppression period of septic patients. Xu et al previously reported that upregulation of RNF146 reduced oxidative stress-induced cell death by inhibiting parthanatos in traumatic brain injury.53 Additionally, studies have found that RNF146 results in the ubiquitination and degradation of phosphatase and tensin homolog which is recognized as a protein substrate of RNF146, to activate the Akt/mTOR pathway and promote tumor cell proliferation and glycolysis.54 Though the relationship of RNF146 and mitochondrial dysfunction and redox imbalance in immune cells has been extensively studied in sepsis, little is known about the function of RNF146 in trained immunity. In this study, RNF146 silencing largely eliminated the role of β-glucan induced training immunity, further confirming the role of RNF146 in the regulation of trained immunity in sepsis. Moreover, our results suggested that the effect of RNF146 in immune trained monocytes/macrophages was carried out through dectin-1-Akt-mTOR activation, and overexpression of RNF146 in RAW264.7 can fine-tune the dectin-1-Akt pathway. All the above results suggested that RNF146 may act as a promising target in sepsis-induced immunosuppression and β-glucan pretreatment induced trained immunity may prevent immunosuppressed state of sepsis.
Our scRNA-seq results also showed that MPA, which is usually regarded as a sensitive measure of platelets activation and the bridge between thrombosis and inflammation,55,56 had an obvious increase in the immunosuppression period of septic patients compared with healthy controls. Recent researches have associated MPA with thrombotic disorders, especially cardiovascular diseases.57–59 Thus, the increase of MPA may be an indicator of hypercoagulable state in immunosuppression state of septic patients. According to the previous researches,60,61 the higher MPA levels are related to higher septic mortality.60 Further studies are needed to identify the role of MPA in sepsis. OxLDL induced trained immunity was reported to acquire inflammatory phenotype of monocyte and result in increased cardiometabolic risk such as atherosclerosis.62 Indeed, epigenetic reprogramming of monocytes induces a long-lasting pro-atherosclerotic phenotype, characterized by increased cytokine and chemokine release, such as IL-1β, which was reported to play a critical role in atherogenesis.63 In Anette’s study, the levels of proinflammatory cytokines IL-1β and IL-6 in oxLDL-induced atherogenesis were two or three times higher than control group,63 while IL-1β and IL-6 levels in β-glucan induced trained immunity model in this study did not exceed twice that of the control group. This suggests a lower risk of atherosclerosis in our model. Furthermore, the survival rate was higher and MSS was lower in β-glucan pretreatment septic mice, suggesting the protective effect of β-glucan in our model. Ganesh et al reported a case that nichi glucan food supplement, which is a 1.3–1,6 β-glucan derived from the black yeast, decreased LDL levels, very low-density lipoprotein levels and triglycerides to the normal range and increased high-density lipoprotein levels in a subject with nondiabetic dyslipidemia on medication with rosuvastatin,64 which also suggesting a possible protective effect against cardiovascular risk of β-glucan. Overall, the only “pre-treatment” beneficial role of β-glucan implies preventive/prophylactic treatment in high-risk septic patients, therefore future study needs to focus on safety in the stimulating trained immunity in sepsis.
Conclusion
This study indicates that β-glucan pretreatment can improve the immunosuppressed state of sepsis, reprogram the metabolism of monocytes and increase the survival rate of septic mice. For the first time, our study demonstrated that E3-ubiquitin ligase RNF146 interacted with dectin-1-Akt pathway in immune trained monocyte/macrophage. The inhibition of dectin-1 by LAM downregulated Akt-RNF146 signaling and partially reversed the β-glucan induced trained immunity in septic mice. These results will help understand the mechanism of trained immunity in sepsis and provide new targets for treating immunosuppression in sepsis.
Data Sharing Statement
The data that support the findings of our study are not publicly available due to their containing information that could compromise the privacy of research participants but are available from the corresponding author JZ upon reasonable request.
Statement of Ethics
The animal procedure, in accordance with the guidelines of the Chinese council on animal care, was approved by the animal review committee of Fudan University Shanghai Cancer Center (FUSCC-IACUC-S2023-0141). Our study was approved by the ethics committee of Zhongshan Hospital Fudan University (B2021-215R) and was registered at the Chinese Clinical Trial Registry (ChiCTR2100048148).
Author Contributions
All authors made a significant contribution to the work reported, whether that is in the conception, study design, execution, acquisition of data, analysis and interpretation, or in all these areas; took part in drafting, revising or critically reviewing the article; gave final approval of the version to be published; have agreed on the journal to which the article has been submitted; and agree to be accountable for all aspects of the work.
Funding
This study was supported by grants from the National Natural Science Foundation of China (No. 82172187, No. 82372164 and No 81971868 to J Zhong; No. 82072213 to CH Miao, Shanghai, China); Shanghai “Yiyuan New Star” Youth Medical Talent Training Funding Program and “Project Achievement” (No. 20234Z0011); Cultivation Project of Medical Teachers (No. DGF828013/022, JG2022-8); Shanghai Clinical Research Center for Anesthesiology (2023ZZ02001).
Disclosure
The authors report no conflicts of interest in this work.
References
1. Cecconi M, Evans L, Levy M, Rhodes A. Sepsis and septic shock. Lancet. 2018;392(10141):75–87.
2. Reinhart K, Daniels R, Kissoon N, Machado FR, Schachter RD, Finfer S. Recognizing Sepsis as a Global Health Priority - A WHO Resolution. New Engl J Med. 2017;377(5):414–417. doi:10.1056/NEJMp1707170
3. Fernando SM, Rochwerg B, Seely AJE. Clinical implications of the Third International Consensus Definitions for Sepsis and Septic Shock (Sepsis-3). CMAJ. 2018;190(36):E1058–e1059. doi:10.1503/cmaj.170149
4. van der Poll T, van de Veerdonk FL, Scicluna BP, Netea MG. The immunopathology of sepsis and potential therapeutic targets. Nat Rev Immunol. 2017;17(7):407–420. doi:10.1038/nri.2017.36
5. Torres LK, Pickkers P, van der Poll T. Tom van der Poll. Sepsis-Induced Immunosuppression. Annu Rev Physiol. 2022;10(84):157–181. doi:10.1146/annurev-physiol-061121-040214
6. Joost Wiersinga W, van der Poll T. Tom van der Poll. Immunopathophysiology of human sepsis. EBioMedicine. 2022;86:104363. doi:10.1016/j.ebiom.2022.104363
7. da Silva Córneo E, Michels M, Dal-Pizzol F. Sepsis, immunosuppression and the role of epigenetic mechanisms. Expert Rev Clin Immunol. 2021;17(2):169–176. doi:10.1080/1744666X.2021.1875820
8. Gustave CA, Gossez M, Demaret J, et al. Septic Shock Shapes B Cell Response toward an Exhausted-like/Immunoregulatory Profile in Patients. J Immunol. 2018;200(7):2418–2425. doi:10.4049/jimmunol.1700929
9. Netea MG, Joosten LA, Latz E, et al. Trained immunity: a program of innate immune memory in health and disease. Science. 2016;352(6284):aaf1098. doi:10.1126/science.aaf1098
10. Tingting P, Shaoqiong S, Yang C, et al. Immune effects of PI3K/Akt/HIF-1α-regulated glycolysis in polymorphonuclear neutrophils during sepsis. Crit Care. 2022;28(26):29.
11. Netea MG, Domínguez-Andrés J, Barreiro LB, et al. Defining trained immunity and its role in health and disease. Nat Rev Immunol. 2020;20(6):375–388. doi:10.1038/s41577-020-0285-6
12. Novakovic B, Habibi E, Wang S-Y, et al. β-Glucan Reverses the Epigenetic State of LPS-Induced Immunological Tolerance. Cell. 2016;167(5):1354–1368.e1314. doi:10.1016/j.cell.2016.09.034
13. Mantovani A, Netea MG, Phimister EG. Trained Innate Immunity, Epigenetics, and Covid-19. New Engl J Med. 2020;383(11):1078–1080. doi:10.1056/NEJMcibr2011679
14. Saeed S, Quintin J, Kerstens HHD, et al. Epigenetic programming of monocyte-to-macrophage differentiation and trained innate immunity. Science. 2014;345(6204):1251086. doi:10.1126/science.1251086
15. Cheng S-C, Quintin J, Cramer RA, et al. mTOR- and HIF-1α–mediated aerobic glycolysis as metabolic basis for trained immunity. Science. 2014;345(6204):1250684. doi:10.1126/science.1250684
16. Singer M, Deutschman CS, Seymour CW, et al. The Third International Consensus Definitions for Sepsis and Septic Shock (Sepsis-3). JAMA. 2016;315(8):801–810. doi:10.1001/jama.2016.0287
17. Reyes M, Vickers D, Billman K, et al. Multiplexed enrichment and genomic profiling of peripheral blood cells reveal subset-specific immune signatures. Sci Adv. 2019;5(1):eaau9223. doi:10.1126/sciadv.aau9223
18. Chen S, Zhou Y, Chen Y, Gu J. fastp: an ultra-fast all-in-one FASTQ preprocessor. Bioinformatics. 2018;34(17):i884–i890. doi:10.1093/bioinformatics/bty560
19. Harriett AJ, Esher Righi S, Lilly EA, et al. Efficacy of candida dubliniensis and fungal β-glucans in inducing trained innate immune protection against inducers of sepsis. Front Cell Infect Microbiol. 2022;12(12):898030. doi:10.3389/fcimb.2022.898030
20. Li N, Zhang Y, Han X, et al. Poly-ADP ribosylation of PTEN by tankyrases promotes PTEN degradation and tumor growth. Genes Dev. 2015;29(2):157–170. doi:10.1101/gad.251785.114
21. Grailer JJ, Fattahi F, Dick RS, Zetoune FS, Ward PA. Cutting edge: critical role for C5aRs in the development of septic lymphopenia in mice. J Immunol. 2015;194(3):868–872. doi:10.4049/jimmunol.1401193
22. Sun Z, Pan Y, Qu J, Xu Y, Dou H, Hou Y. 17β-Estradiol Promotes Trained Immunity in Females Against Sepsis via Regulating Nucleus Translocation of RelB. Front Immunol. 2020;11:1591. doi:10.3389/fimmu.2020.01591
23. Kim H, Park J, Kang H, et al. Activation of the Akt1-CREB pathway promotes RNF146 expression to inhibit PARP1-mediated neuronal death. Sci Signaling. 2020;13(663). doi:10.1126/scisignal.aax7119
24. Pan T, Sun S, Chen Y, et al. Immune effects of PI3K/Akt/HIF-1α-regulated glycolysis in polymorphonuclear neutrophils during sepsis. Critical Care. 2022;26(1):29. doi:10.1186/s13054-022-03893-6
25. Hillmer EJ, Zhang H, Li HS, Watowich SS. STAT3 signaling in immunity. Cytokine Growth Factor Rev. 2016;9(31):1–15. doi:10.1016/j.cytogfr.2016.05.001
26. Heng Y, Zhang X, Borggrewe M, et al. Systemic administration of β-glucan induces immune training in microglia. J Neuroinflammation. 2021;18(1):57. doi:10.1186/s12974-021-02103-4
27. Eljaszewicz A, Ruchti F, Radzikowska U, et al. Trained immunity and tolerance in innate lymphoid cells, monocytes, and dendritic cells during allergen-specific immunotherapy. J Allergy Clin Immunol. 2021;147(5):1865–1877. doi:10.1016/j.jaci.2020.08.042
28. Kawamura S, Ohteki T. Monopoiesis in humans and mice. Intern immun. 2018;30(11):503–509. doi:10.1093/intimm/dxy063
29. Hajishengallis G, Li X, Mitroulis I, Chavakis T. Trained Innate Immunity and Its Implications for Mucosal Immunity and Inflammation. Adv Exp Med Biol. 2019;1197:11–26.
30. Kang HC, Lee YI, Shin JH, et al. Iduna is a poly(ADP-ribose) (PAR)-dependent E3 ubiquitin ligase that regulates DNA damage.
31. Zhou ZD, Chan CH, Xiao ZC, Tan EK. Ring finger protein 146/Iduna is a poly(ADP-ribose) polymer binding and PARsylation dependent E3 ubiquitin ligase. Cell Adh Migr. 2011;5(6):463–471. doi:10.4161/cam.5.6.18356
32. Roquilly A, McWilliam HEG, Jacqueline C, et al. Local Modulation of Antigen-Presenting Cell Development after Resolution of Pneumonia Induces Long-Term Susceptibility to Secondary Infections. Immunity. 2017;47(1):135–147.e135. doi:10.1016/j.immuni.2017.06.021
33. Carson WF, Cavassani KA, Dou Y, Kunkel SL. Epigenetic regulation of immune cell functions during post-septic immunosuppression. Epigenetics. 2011;6(3):273–283. doi:10.4161/epi.6.3.14017
34. Qiu P, Liu Y, Zhang J. Review: the Role and Mechanisms of Macrophage Autophagy in Sepsis. Inflammation. 2019;42(1):6–19. doi:10.1007/s10753-018-0890-8
35. Hotchkiss RS, Monneret G, Payen D. Sepsis-induced immunosuppression: from cellular dysfunctions to immunotherapy. Nat Rev Immunol. 2013;13(12):862–874. doi:10.1038/nri3552
36. Nascimento DC, Alves-Filho JC, Sônego F, et al. Role of regulatory T cells in long-term immune dysfunction associated with severe sepsis. Crit Care Med. 2010;38(8):1718–1725. doi:10.1097/CCM.0b013e3181e78ad0
37. Muenzer JT, Davis CG, Dunne BS, Unsinger J, Dunne WM, Hotchkiss RS. Pneumonia after cecal ligation and puncture: a clinically relevant ”two-hit” model of sepsis. Shock Augusta ga. 2006;26(6):565–570. doi:10.1097/01.shk.0000235130.82363.ed
38. Rodriguez S, Chora A, Goumnerov B, et al. Dysfunctional expansion of hematopoietic stem cells and block of myeloid differentiation in lethal sepsis. Blood. 2009;114(19):4064–4076. doi:10.1182/blood-2009-04-214916
39. McBride MA, Owen AM, Stothers CL, et al. The Metabolic Basis of Immune Dysfunction Following Sepsis and Trauma. Front Immunol. 2020;11:1043. doi:10.3389/fimmu.2020.01043
40. Bomans K, Schenz J, Sztwiertnia I, Schaack D, Weigand MA, Uhle F. Sepsis Induces a Long-Lasting State of Trained Immunity in Bone Marrow Monocytes. Front Immunol. 2018;9:2685. doi:10.3389/fimmu.2018.02685
41. Raghavan K, Dedeepiya VD, Suryaprakash V, et al. Beneficial effects of novel aureobasidium pullulans strains produced beta-1,3-1,6 glucans on interleukin-6 and D-dimer levels in COVID-19 patients; results of a randomized multiple-arm pilot clinical study. Biomed Pharmacothe. 2022;145:112243. doi:10.1016/j.biopha.2021.112243
42. Vetvicka V, Vannucci L, Sima P, Richter J. Beta Glucan: supplement or Drug? From Laboratory to Clinical Trials. Molecules. 2019;24(7):1251. doi:10.3390/molecules24071251
43. Preethy S, Raghavan K, Dedeepiya VD, et al. Beneficial Immune Regulation by Biological Response Modifier Glucans in COVID-19 and Their Envisaged Potentials in the Management of Sepsis. Front Immunol. 2022;13:870632. doi:10.3389/fimmu.2022.870632
44. Tannahill GM, Curtis AM, Adamik J, et al. Succinate is a danger signal that induces IL-1β via HIF-1α. Nature. 2013;496(7444):238–242. doi:10.1038/nature11986
45. Arts RJ, Novakovic B, Ter Horst R, et al. Glutaminolysis and Fumarate Accumulation Integrate Immunometabolic and Epigenetic Programs in Trained Immunity. Cell Metab. 2016;24(6):807–819. doi:10.1016/j.cmet.2016.10.008
46. Jung J, Zeng H, Horng T. Metabolism as a guiding force for immunity. Nat Cell Biol. 2019;21(1):85–93. doi:10.1038/s41556-018-0217-x
47. de Baat A, Trinh B, Ellingsgaard H, Donath MY. Physiological role of cytokines in the regulation of mammalian metabolism. Trends Immunol. 2023;44(8):613–627. doi:10.1016/j.it.2023.06.002
48. Bahoosh SR, Shokoohinia Y, Eftekhari M. Glucosinolates and their hydrolysis products as potential nutraceuticals to combat cytokine storm in SARS-CoV-2. DARU J Pharm Sci. 2022;30:245–252. doi:10.1007/s40199-022-00435-x
49. Quintin J, Saeed S, Martens JHA, et al. Candida albicans infection affords protection against reinfection via functional reprogramming of monocytes. Cell Host Microbe. 2012;12(2):223–232. doi:10.1016/j.chom.2012.06.006
50. Mitroulis I, Ruppova K, Wang B, et al. Modulation of Myelopoiesis Progenitors Is an Integral Component of Trained Immunity. Cell. 2018;172(1–2):147–161.e112. doi:10.1016/j.cell.2017.11.034
51. Mata-Martínez P, Bergón-Gutiérrez M, Del Fresno C. Dectin-1 Signaling Update: new Perspectives for Trained Immunity. Front Immunol. 2022;13:812148. doi:10.3389/fimmu.2022.812148
52. Krawczyk CM, Holowka T, Sun J, et al. Toll-like receptor-induced changes in glycolytic metabolism regulate dendritic cell activation. Blood. 2010;115(23):4742–4749. doi:10.1182/blood-2009-10-249540
53. Xu H, Li X, Wu X, et al. Iduna protects HT22 cells by inhibiting parthanatos: the role of the p53-MDM2 pathway. Exp. Cell. Res. 2019;384(1):111547. doi:10.1016/j.yexcr.2019.111547
54. Shen G, Wang H, Zhu N, et al. HIF-1/2α-Activated RNF146 Enhances the Proliferation and Glycolysis of Hepatocellular Carcinoma Cells via the PTEN/AKT/mTOR Pathway. Front Cell Develop Biol. 2022;10:893888. doi:10.3389/fcell.2022.893888
55. Michelson AD, Barnard MR, Krueger LA, Valeri CR, Furman MI. Circulating monocyte-platelet aggregates are a more sensitive marker of in vivo platelet activation than platelet surface P-selectin: studies in baboons, human coronary intervention, and human acute myocardial infarction. Circulation. 2001;104(13):1533–1537. doi:10.1161/hc3801.095588
56. Freedman JE, Loscalzo J. Platelet-monocyte aggregates: bridging thrombosis and inflammation. Circulation. 2002;105(18):2130–2132. doi:10.1161/01.CIR.0000017140.26466.F5
57. Rolling CC, Barrett TJ, Berger JS. Platelet-monocyte aggregates: molecular mediators of thromboinflammation. Frontiers in Cardiovascular Medicine. 2023;10:960398. doi:10.3389/fcvm.2023.960398
58. Stojkovic S, Wadowski PP, Haider P, et al. Circulating MicroRNAs and Monocyte-Platelet Aggregate Formation in Acute Coronary Syndrome. Thromb Haemos. 2021;121(7):913–922. doi:10.1055/s-0040-1722226
59. Kamińska J, Lisowska A, Koper-Lenkiewicz OM, et al. Differences in Monocyte Subsets and Monocyte-Platelet Aggregates in Acute Myocardial Infarction-PreliminaryResults. Am J Med Sci. 2019;357(5):421–434. doi:10.1016/j.amjms.2019.02.010
60. Fu G, Deng M, Neal MD, Billiar TR, Scott MJ. Platelet-Monocyte Aggregates: understanding Mechanisms and Functions in Sepsis. Shock Augusta ga. 2021;55(2):156–166. doi:10.1097/SHK.0000000000001619
61. Wu Q, Ren J, Hu D, et al. Monocyte subsets and monocyte-platelet aggregates: implications in predicting septic mortality among surgical critical illness patients. Biomarkers. 2016;21(6):509–516. doi:10.3109/1354750X.2016.1160290
62. Christ A, Günther P, LNetea MG, et al. Western diet triggers NLRP3-dependent innate immune reprogramming. Cell. 2018;172:162–175. doi:10.1016/j.cell.2017.12.013
63. Bekkering S, Quintin J, Netea MG, et al. Oxidized low-density lipoprotein induces long-term proinflammatory cytokine production and foam cell formation via epigenetic reprogramming of monocytes. Arterioscler Thromb Vasc Biol. 2014;34:1731–1738. doi:10.1161/ATVBAHA.114.303887
64. Ganeh JS, Rao YY, Ravikumar R, et al. Beneficial effects of black yeast derived 1-3, 1-6 beta glucan-nichi glucan in a dyslipidemic individual of Indian origin- a case report. J Dietary Suppl. 2014;11(1):1–6. doi:10.3109/19390211.2013.859211
© 2025 The Author(s). This work is published and licensed by Dove Medical Press Limited. The
full terms of this license are available at https://www.dovepress.com/terms.php
and incorporate the Creative Commons Attribution
- Non Commercial (unported, 3.0) License.
By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted
without any further permission from Dove Medical Press Limited, provided the work is properly
attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.
Recommended articles

Advances in the Study of Immunosuppressive Mechanisms in Sepsis
Fu X, Liu Z, Wang Y
Journal of Inflammation Research 2023, 16:3967-3981
Published Date: 8 September 2023