1.Itaconate derivative 4-OI inhibits M1 macrophage polarization and restores its impaired function in immune thrombocytopenia through metabolic reprogramming.
Qiang LIU ; Anli LIU ; Shaoqiu LENG ; Xiaoyu ZHANG ; Xiaolin WANG ; Zhang CHENG ; Shuwen WANG ; Jun PENG ; Qi FENG
Chinese Medical Journal 2025;138(16):2006-2015
BACKGROUND:
Macrophage polarization anomalies and dysfunction play a crucial role in the pathogenesis of immune thrombocytopenia (ITP). Itaconate is a Krebs cycle-derived immunometabolite synthesized by myeloid cells to modulate cellular metabolism and inflammatory responses. This study aimed to evaluate the immunoregulatory effects of an itaconate derivative on macrophages in patients with ITP.
METHODS:
Peripheral blood-derived macrophages from patients with ITP and healthy controls were treated with 4-octyl itaconate (4-OI), a derivative of itaconate that can penetrate the cell membrane. Macrophage polarization, antigen-presenting functions, and phagocytic capability were measured via flow cytometry and enzyme-linked immunosorbent assay (ELISA). Macrophage glycolysis in patients with ITP and the metabolic regulatory effect of 4-OI were detected using a Seahorse XFe96 Analyzer. An active murine model of ITP was used to evaluate the therapeutic effects of 4-OI in vivo .
RESULTS:
4-OI reduced the levels of CD80 and CD86 in M1 macrophages and suppressed the release of tumor necrosis factor-α (TNF-α) and interleukin (IL)-6 pro-inflammatory cytokines, suggesting that 4-OI could hinder the polarization of macrophages toward an M1 phenotype. We found that 4-OI pretreated M1 macrophages reduced the proliferation of CD4 + T cells and promoted the differentiation of regulatory T cells. In addition, after 4-OI treatment, the phagocytic capacity of M1 macrophages toward antibody-coated platelets decreased significantly in patients with ITP. In addition, the glycolytic function of M1 macrophages was elevated in individuals with ITP compared to those in healthy controls. 4-OI treatment downregulated glycolysis in M1 macrophages. The glycolysis inhibitor 2-deoxy-d-glucose (2-DG) also inhibited the polarization of M1 macrophages and restored their functions. In vivo , 4-OI treatment significantly increased platelet counts in the active ITP murine model.
CONCLUSIONS
Itaconate derivative 4-OI inhibited M1 macrophage polarization and restored impaired functions through metabolic reprogramming. This study provides a novel therapeutic option for ITP.
Macrophages/metabolism*
;
Humans
;
Animals
;
Succinates/pharmacology*
;
Mice
;
Male
;
Female
;
Adult
;
Middle Aged
;
Flow Cytometry
;
Tumor Necrosis Factor-alpha/metabolism*
;
Enzyme-Linked Immunosorbent Assay
;
Purpura, Thrombocytopenic, Idiopathic/metabolism*
;
Glycolysis/drug effects*
;
Metabolic Reprogramming
2.Mechanism of total flavone of Abelmoschus manihot in treating ulcerative colitis and depression via intestinal flora-glycerophospholipid metabolism- macrophage polarization pathway.
Chang-Ye LU ; Xiao-Min YUAN ; Lin-Hai HE ; Jia-Rong MAO ; Yu-Gen CHEN
China Journal of Chinese Materia Medica 2025;50(5):1286-1297
This study delves into the mechanism of total flavone of Abelmoschus manihot(TFA) in treating ulcerative colitis(UC) and depression via inhibiting M1 polarization of macrophages and reshaping intestinal flora and glycerolphospholipid metabolism. The study established a mouse model of UC and depression induced by chronic restraint stress(CRS) and dextran sulfate sodium(DSS). The fecal microbiota transplantation(FMT) experiment after TFA intervention was conducted. Mice in the FMT donor group were modeled and treated, and fecal samples were taken to prepare the bacterial solution. Mice in the FMT receptor group were treated with antibiotic intervention, and then administered bacterial solution by gavage from mice in the donor group, followed by UC depression modeling. After the experiment, behavioral tests were conducted to evaluate depressive-like behaviors by measuring the levels of 5-hydroxytryptamine(5-HT) and brain-derived neurotrophic factor(BDNF) in the hippocampus of mice. The levels of tumor necrosis factor-α(TNF-α),interleukin-6(IL-6),and interleukin-1β(IL-1β)in the brain and colon tissue of mice were also measured, and the polarization status of macrophages was evaluated by measuring the mRNA levels of CD86 and CD206. 16S ribosomal RNA(16S rRNA) sequencing technology was used to analyze changes in the intestinal flora of mice. Wide target lipidomics was used to detect serum lipid metabolite levels in mice after FMT,and correlation analysis was conducted between lipids and differential intestinal flora significantly regulated by TFA. In vitro experiments, representative glycerophospholipid metabolites and glycerophospholipid inhibitors were used to intervene in Raw264.7 macrophages, and the mRNA levels of TNF-α,IL-6,IL-1β,CD86,and CD206 were detected. The results showed that TFA and FMT after intervention could significantly improve depressive-like behavior and intestinal inflammation in mice with UC and depression, significantly downregulate pro-inflammatory cytokines and CD86 mRNA expression in brain and colon tissue, inhibiting M1 polarization of macrophages, and significantly upregulate CD206 mRNA expression, promoting M2 polarization of macrophages. In addition, the high-dose group had a more significant effect. After TFA intervention, FMT significantly corrected the metabolic disorder of glycerophospholipids in mice with UC and depression, and there was a significant correlation between differential intestinal flora and glycerophospholipids. In vitro experiments showed that glycerophospholipid metabolites, especially lysophosphatidylcholine(LPC),significantly upregulated pro-inflammatory cytokines and CD86 mRNA expression, promote M1 polarization of macrophages, while glycerophospholipid inhibitors had the opposite effect. The results indicate that TFA effectively treats depression and UC by correcting intestinal flora dysbiosis and reshaping glycerophospholipid metabolism, thereby inhibiting M1 polarization of macrophages.
Animals
;
Mice
;
Gastrointestinal Microbiome/drug effects*
;
Abelmoschus/chemistry*
;
Macrophages/metabolism*
;
Colitis, Ulcerative/immunology*
;
Flavones/administration & dosage*
;
Male
;
Depression/genetics*
;
Glycerophospholipids/metabolism*
;
Humans
;
Drugs, Chinese Herbal/administration & dosage*
;
Mice, Inbred C57BL
3.Study on protective effect of arbutin in yam on acute lung injury and its metabolic regulation mechanism.
Kai-Li YE ; Meng-Nan ZENG ; Feng-Xiao HAO ; Peng-Li GUO ; Yu-Han ZHANG ; Wei-Sheng FENG ; Xiao-Ke ZHENG
China Journal of Chinese Materia Medica 2025;50(15):4100-4109
This study investigated the protective effect of arbutin(Arb) in yam on lipopolysaccharide(LPS)-induced acute lung injury(ALI) in a mouse model and revealed its possible mechanism of action by metabolomics technology, providing a theoretical basis for clinical treatment of ALI. SPF BALB/c mice were randomly divided into normal control group, model group, resveratrol(Rv)-positive control group, Arb low-dose(15 mg·kg~(-1)) group, and Arb high-dose(30 mg·kg~(-1)) group. The LPS-induced ALI model was established in all groups except the normal control group. Hematoxylin-eosin(HE) staining, TUNEL staining, and WBP whole-body non-invasive pulmonary function testing were used to evaluate the degree of lung tissue damage and lung function changes. Enzyme-linked immunosorbent assay(ELISA) was used to detect the level of inflammatory factors in lung tissue. Flow cytometry was used to analyze the M1/M2 polarization status of macrophages in lung tissue. Western blot was used to detect the expression levels of the TLR4 signaling pathway and related apoptotic proteins. Liquid chromatograph-mass spectrometer(LC-MS) metabolomics was used to analyze the changes in serum metabolic profile after Arb intervention. The results showed that Arb pretreatment significantly alleviated LPS-induced lung tissue injury, improved lung function, reduced the levels of pro-inflammatory factors(IL-6, TNF-α, IL-18, and IL-1β), and regulated the polarization status of M1/M2 macrophages. In addition, Arb inhibited the activation of the TLR4 signaling pathway, reduced the expression of pro-apoptotic proteins such as Bax, caspase-3, and caspase-9, up-regulated the level of Bcl-2 protein, and inhibited apoptosis of lung cells. Metabolomic analysis showed that Arb significantly improved LPS-induced metabolic abnormalities, mainly involving key pathways such as galactose metabolism, phenylalanine metabolism, and lipid metabolism. In summary, Arb can significantly reduce LPS-induced ALI by regulating the release of inflammatory factors, inhibiting the activation of the TLR4 signaling pathway, improving metabolic disorders, and regulating macrophage polarization, indicating that Arb has potential clinical application value.
Animals
;
Acute Lung Injury/chemically induced*
;
Mice
;
Mice, Inbred BALB C
;
Arbutin/administration & dosage*
;
Male
;
Toll-Like Receptor 4/immunology*
;
Apoptosis/drug effects*
;
Lung/metabolism*
;
Signal Transduction/drug effects*
;
Protective Agents/administration & dosage*
;
Humans
;
Macrophages/immunology*
;
Drugs, Chinese Herbal/administration & dosage*
4.Molecular mechanism of magnesium alloy promoting macrophage M2 polarization through modulation of PI3K/AKT signaling pathway for tendon-bone healing in rotator cuff injury repair.
Xianhao SHENG ; Wen ZHANG ; Shoulong SONG ; Fei ZHANG ; Baoxiang ZHANG ; Xiaoying TIAN ; Wentao XIONG ; Yingguang ZHU ; Yuxin XIE ; Zi'ang LI ; Lili TAN ; Qiang ZHANG ; Yan WANG
Chinese Journal of Reparative and Reconstructive Surgery 2025;39(2):174-186
OBJECTIVE:
To evaluate the effect of biodegradable magnesium alloy materials in promoting tendon-bone healing during rotator cuff tear repair and to investigate their potential underlying biological mechanisms.
METHODS:
Forty-eight 8-week-old Sprague Dawley rats were taken and randomly divided into groups A, B, and C. Rotator cuff tear models were created and repaired using magnesium alloy sutures in group A and Vicryl Plus 4-0 absorbable sutures in group B, while only subcutaneous incisions and sutures were performed in group C. Organ samples of groups A and B were taken for HE staining at 1 and 2 weeks after operation to evaluate the safety of magnesium alloy, and specimens from the supraspinatus tendon and proximal humerus were harvested at 2, 4, 8, and 12 weeks after operation. The specimens were observed macroscopically at 4 and 12 weeks after operation. Biomechanical tests were performed at 4, 8, and 12 weeks to test the ultimate load and stiffness of the healing sites in groups A and B. At 2, 4, and 12 weeks, the specimens were subjected to the following tests: Micro-CT to evaluate the formation of bone tunnels in groups A and B, HE staining and Masson staining to observe the regeneration of fibrocartilage at the tendon-bone interface after decalcification and sectioning, and Goldner trichrome staining to evaluate the calcification. Immunohistochemical staining was performed to detect the expressions of angiogenic factors, including vascular endothelial growth factor (VEGF) and bone morphogenetic protein 2 (BMP-2), as well as osteogenic factors at the tendon-bone interface. Additionally, immunofluorescence staining was used to examine the expressions of Arginase 1 and Integrin beta-2 to assess M1 and M2 macrophage polarization at the tendon-bone interface. The role of the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT) signaling pathway in tendon-bone healing was further analyzed using real-time fluorescence quantitative PCR.
RESULTS:
Analysis of visceral sections revealed that magnesium ions released during the degradation of magnesium alloys did not cause significant toxic effects on organs such as the heart, liver, spleen, lungs, and kidneys, indicating good biosafety. Histological analysis further demonstrated that fibrocartilage regeneration at the tendon-bone interface in group A occurred earlier, and the amount of fibrocartilage was significantly greater compared to group B, suggesting a positive effect of magnesium alloy material on tendon-bone interface repair. Additionally, Micro-CT analysis results revealed that bone tunnel formation occurred more rapidly in group A compared to group B, further supporting the beneficial effect of magnesium alloy on bone healing. Biomechanical testing showed that the ultimate load in group A was consistently higher than in group B, and the stiffness of group A was also greater than that of group B at 4 weeks, indicating stronger tissue-carrying capacity following tendon-bone interface repair and highlighting the potential of magnesium alloy in enhancing tendon-bone healing. Immunohistochemical staining results indicated that the expressions of VEGF and BMP-2 were significantly upregulated during the early stages of healing, suggesting that magnesium alloy effectively promoted angiogenesis and bone formation, thereby accelerating the tendon-bone healing process. Immunofluorescence staining further revealed that magnesium ions exerted significant anti-inflammatory effects by regulating macrophage polarization, promoting their shift toward the M2 phenotype. Real-time fluorescence quantitative PCR results demonstrated that magnesium ions could facilitate tendon-bone healing by modulating the PI3K/AKT signaling pathway.
CONCLUSION
Biodegradable magnesium alloy material accelerated fibrocartilage regeneration and calcification at the tendon-bone interface in rat rotator cuff tear repair by regulating the PI3K/AKT signaling pathway, thereby significantly enhancing tendon-bone healing.
Animals
;
Rotator Cuff Injuries/metabolism*
;
Rats, Sprague-Dawley
;
Signal Transduction
;
Wound Healing/drug effects*
;
Alloys/pharmacology*
;
Rats
;
Proto-Oncogene Proteins c-akt/metabolism*
;
Rotator Cuff/metabolism*
;
Macrophages/metabolism*
;
Magnesium/pharmacology*
;
Phosphatidylinositol 3-Kinases/metabolism*
;
Vascular Endothelial Growth Factor A/metabolism*
;
Male
;
Biocompatible Materials
;
Bone Morphogenetic Protein 2/metabolism*
5.Astragaloside IV regulates Snail1 lactylation and acetylation to mediate macrophage polarization and improve myocardial infarction.
Shaopeng CHEN ; Rudian KANG ; Xinbao HONG ; Yilong LIU
Chinese Journal of Cellular and Molecular Immunology 2025;41(4):289-299
Objective To investigate the impact of Astragaloside-IV (AS-IV) on the progression of myocardial infarction (MI) through macrophage-dependent mechanisms by regulating Snail1 lactylation and acetylation, as well as the transforming growth factor β (TGF-β) pathway. Methods Oxygen glucose deprivation (OGD) was used to establish an in vitro myocardial ischemia model in rat cardiomyocytes (H9c2), which were then treated with AS-IV. Cell viability was assessed using CCK-8, apoptosis was evaluated by flow cytometry, and LDH levels were measured to assess cellular damage. RAW246.7 cells were treated with LPS, and lactate levels in the supernatant were measured using ELISA, while expression of macrophage phenotype markers was evaluated using Western blot. RAW246.7 cell-conditioned medium (CM) was co-cultured with H9c2 cells to assess the protective effects of AS-IV on macrophage CM-mediated H9c2 damage. RAW246.7 cells were induced to differentiate into M1-like macrophages using LPS (100 ng/mL) + IFN-γ (20 ng/mL), and Snail1 was overexpressed in M1 macrophages. Transfected M1 macrophage CM was co-cultured with H9c2 cells to validate the mechanisms of AS-IV in MI. An MI rat model was established by ligation of the left anterior descending coronary artery (LAD), and was treated with AS-IV. Cardiac function, myocardial cell apoptosis, and cardiac tissue pathology were studied using echocardiography, TUNEL, and HE staining, respectively. Results Compared to the OGD group, AS-IV treatment promoted cell viability, reduced apoptosis and decreased LDH release. LPS upregulated lactate levels in the supernatant of RAW246.7 cell cultures and induced polarization of RAW246.7 cells to the M1 phenotype. AS-IV attenuated the damaging effects of RAW246.7 cell CM on H9c2 cells . Overexpression of Snail1 in M1 macrophages weakened the protective effects of AS-IV on H9c2 cells . In vivo study, results showed that, compared to the MI group, AS-IV treatment reduced lactate levels in the hearts of MI rats, improved cardiac function and myocardial injury and attenuated myocardial cell apoptosis. Conclusion AS-IV inhibits TGF-β pathway activation through the suppression of Snail1 lactylation and acetylation in a macrophage-dependent manner, thereby mitigating myocardial cell damage following MI.
Animals
;
Myocardial Infarction/drug therapy*
;
Rats
;
Snail Family Transcription Factors/metabolism*
;
Macrophages/cytology*
;
Myocytes, Cardiac/metabolism*
;
Triterpenes/pharmacology*
;
Saponins/pharmacology*
;
Acetylation/drug effects*
;
Apoptosis/drug effects*
;
Mice
;
Cell Line
;
RAW 264.7 Cells
;
Transforming Growth Factor beta/metabolism*
6.Research on the mechanism of gentiopicroside preventing macrophage-mediated liver fibrosis by regulating the MIF-SPP1 signaling pathway in hepatic stellate cells.
Jixu WANG ; Yingbin ZHU ; Maoli CHEN ; Yongfeng HAN
Chinese Journal of Cellular and Molecular Immunology 2025;41(7):593-602
Objective To explore the mechanism by which gentiopicroside (GPS) prevents macrophage-mediated hepatic fibrosis by regulating the macrophage migration inhibitory factor (MIF)-secreted phosphoprotein 1 (SPP1) signaling pathway in hepatic stellate cells. Methods LX-2 cells were divided into control group, transforming growth factor β(TGF-β) group, and TGF-β combined with GPS (25, 50, 100, 150 μmol/mL) groups. Cell proliferation was detected by EDU assay, cell invasion was assessed by TranswellTM assay, and the protein expressions of α-smooth muscle actin (α-SMA) and type I collagen (COL1A1) were measured by Western blot. M1-type macrophage-conditioned medium (M1-CM) was used to treat LX-2 cells in the TGF-β group and TGF-β combined with GPS group. The concentrations of inducible nitric oxide synthase (iNOS) and arginase 1 (Arg1) in the cell supernatant, as well as cell proliferation, invasion ability, and the expressions of α-SMA and COL1A1 were detected. Bioinformatics analysis was performed to identify the target intersections of GPS, hepatic fibrosis, and macrophage-related genes. Drug affinity responsive target stability (DARTS) experiments and Western blot were used to verify the regulatory effect of GPS on MIF. Furthermore, LX-2 cells were divided into control group, TGF-β group, TGF-β combined with M2-CM group, TGF-β and oe-NC combined with M2-CM group, and TGF-β and oe-MIF combined with M2-CM group to analyze the concentrations of iNOS and Arg1 in the cell supernatant, as well as changes in cell proliferation, invasion, and the expressions of α-SMA and COL1A1. LX-2 cells were also divided into control group, TGF-β group, TGF-β combined with oe-NC group, TGF-β combined with oe-MIF group, and TGF-β and oe-MIF combined with GPS group to determine the protein expressions of MIF and SPP1 by Western blot. A rat model of hepatic fibrosis was constructed to explore the potential therapeutic effects of GPS on hepatic fibrosis in vivo. Results Compared with the control group, the proliferation and invasion abilities of LX-2 cells in the TGF-β group were increased, and the protein expressions of α-SMA and COL1A1 were enhanced. GPS intervention inhibited the proliferation and invasion of LX-2 cells under TGF-β conditions and reduced the expressions of α-SMA and COL1A1. Compared with the control group, the concentration of iNOS in the cell supernatant of the TGF-β group was upregulated, while the concentration of Arg1 was decreased. M1-CM treatment further increased the concentration of iNOS, decreased the concentration of Arg1, and promoted cell proliferation and invasion, as well as upregulated the expressions of α-SMA and COL1A1 on the basis of TGF-β intervention. However, GPS could reverse the effects of M1-CM intervention. Bioinformatics analysis revealed that MIF was one of the target intersections of GPS, hepatic fibrosis, and macrophage-related genes, and GPS could target and inhibit its expression. Compared with the TGF-β group, after M2-CM intervention, the concentration of iNOS in the cell supernatant decreased, the concentration of Arg1 increased, the proliferation and invasion abilities of LX-2 cells were reduced, and the expressions of α-SMA and COL1A1 were weakened. However, overexpression of MIF reversed the effects of M2-CM intervention. Western blot results showed that compared with the control group, the protein expressions of MIF and SPP1 were enhanced in the TGF-β group. Overexpression of MIF further enhanced the expressions of MIF and SPP1, while GPS intervention inhibited the expressions of MIF and SPP1. In the animal experiment, GPS intervention treatment alleviated liver injury in rats with hepatic fibrosis and inhibited the expressions of MIF and SPP1, as well as α-SMA and COL1A1 in liver tissue. Conclusion GPS may prevent macrophage-mediated hepatic fibrosis by inhibiting the MIF-SPP1 signaling pathway in hepatic stellate cells.
Hepatic Stellate Cells/metabolism*
;
Signal Transduction/drug effects*
;
Macrophage Migration-Inhibitory Factors/genetics*
;
Liver Cirrhosis/prevention & control*
;
Macrophages/drug effects*
;
Iridoid Glucosides/pharmacology*
;
Humans
;
Cell Proliferation/drug effects*
;
Animals
;
Cell Line
;
Collagen Type I/metabolism*
;
Collagen Type I, alpha 1 Chain
;
Intramolecular Oxidoreductases/genetics*
;
Rats
;
Transforming Growth Factor beta/pharmacology*
;
Actins/metabolism*
7.PM2.5-induced M2 Polarization and IL-1α Secretion by Tumor-associated Macrophages Promotes Lung Adenocarcinoma Progression.
Bomiao QING ; Xiaolan LI ; Qin RAN ; Guoping LI
Chinese Journal of Lung Cancer 2025;28(9):667-679
BACKGROUND:
Lung adenocarcinoma (LUAD) remains one of the leading causes of cancer morbidity and mortality worldwide, and its initiation and progression are closely associated with the tumor immune microenvironment. Increasing evidence suggests that environmental exposure is a critical factor influencing lung cancer development. Among these factors, fine particulate matter (PM2.5), a major component of air pollution, has been strongly linked to elevated lung cancer risk and unfavorable prognosis. However, the underlying immunoregulatory mechanisms by which PM2.5 drives LUAD progression remain poorly understood. Tumor-associated macrophages (TAMs), especially those polarized toward the M2 phenotype, are key components of the tumor microenvironment and play crucial roles in tumor growth, angiogenesis, and immune evasion. This study aims to investigate the effects of PM2.5 exposure on TAMs and to identify the key pro-tumorigenic factors mediating this process.
METHODS:
A mouse orthotopic lung cancer model under PM2.5 exposure was established to assess lung tumor growth and macrophage phenotypic alterations using in vivo imaging and flow cytometry. A subcutaneous tumor model involving co-inoculated macrophages and tumor cells was used to further verify the effects of PM2.5 on the function of TAMs and tumor malignancy. Combining in vitro experiments, flow cytometry, Western blot, reverse transcription quantitative polymerase chain reaction (RT-qPCR), cell counting kit-8 (CCK-8) assay, colony formation assay, and wound healing assay were employed to evaluate the regulatory effects of PM2.5 on the polarization of bone marrow-derived macrophages (BMDMs) as well as tumor cell proliferation, migration, and colony-forming ability. Transcriptome sequencing integrated with TISIDB (Tumor-immune System Interactions Database) and GEPIA (Gene Expression Profiling Interactive Analysis) databases was performed to identify key cytokines for further functional validation.
RESULTS:
In the mouse orthotopic lung cancer model, PM2.5 exposure significantly promoted tumor growth and increased the proportion of M2-type TAMs (P<0.05). Subcutaneous co-inoculation with PM2.5-treated BMDMs markedly enhanced tumor proliferation and elevated the intratumoral M2-type TAMs. PM2.5-pretreated BMDMs exhibited an immunosuppressive programmed cell death ligand 1 (PD-L1)+/arginase 1 (Arg1)+ phenotype, and their conditioned media significantly promoted proliferation, migration, and colony formation of Lewis lung carcinoma cells (LLC) and B16 melanoma cells (B16) (P<0.05). Transcriptome analysis revealed that PM2.5 substantially altered macrophage gene expression, with IL-1α identified as a key upregulated secreted cytokine enriched in immunosuppressive related signaling pathways. Clinical database analyses further indicated that IL-1α expression was positively correlated with macrophage and regulatory T cells (Treg) infiltration in the LUAD immune microenvironment, and that high IL-1α expression was associated with worse overall survival in LUAD patients (HR=1.5, P=0.0053). Western blot, RT-qPCR, and immunofluorescence confirmed that PM2.5 exposure significantly upregulated IL-1α expression and secretion in TAMs.
CONCLUSIONS
PM2.5 exposure facilitates LUAD progression by inducing an immunosuppressive phenotype in macrophages and enhancing the malignant behaviors of tumor cells. Mechanistically, IL-1α may serve as a key pro-tumorigenic cytokine secreted by macrophages under PM2.5 exposure. This study provides new insights into the pathogenesis of PM2.5-associated LUAD and suggests that IL-1α could serve as a potential therapeutic target.
Animals
;
Mice
;
Tumor-Associated Macrophages/immunology*
;
Particulate Matter/toxicity*
;
Adenocarcinoma of Lung/metabolism*
;
Lung Neoplasms/genetics*
;
Humans
;
Disease Progression
;
Tumor Microenvironment/drug effects*
;
Cell Proliferation/drug effects*
;
Cell Line, Tumor
8.Tanreqing Injection Inhibits Activation of NLRP3 Inflammasome in Macrophages Infected with Influenza A Virus by Promoting Mitophagy.
Tian-Yi LIU ; Yu HAO ; Qin MAO ; Na ZHOU ; Meng-Hua LIU ; Jun WU ; Yi WANG ; Ming-Rui YANG
Chinese journal of integrative medicine 2025;31(1):19-27
OBJECTIVE:
To investigate the inhibitory effect of Tanreqing Injection (TRQ) on the activation of nucleotide-binding oligomerization domain-like receptor pyrin domain containing 3 (NLRP3) inflammasome in macrophages infected with influenza A virus and the underlying mechanism based on mitophagy pathway.
METHODS:
The inflammatory model of murine macrophage J774A.1 induced by influenza A virus [strain A/Puerto Rico/8/1934 (H1N1), PR8] was constructed and treated by TRQ, while the mitochondria-targeted antioxidant Mito-TEMPO and autophagy specific inhibitor 3-methyladenine (3-MA) were used as controls to intensively study the anti-inflammatory mechanism of TRQ based on mitophagy-mitochondrial reactive oxygen species (mtROS)-NLRP3 inflammasome pathway. The levels of NLRP3, Caspase-1 p20, microtubule-associated protein 1 light chain 3 II (LC3II) and P62 proteins were measured by Western blot. The release of interleukin-1β (IL-1β) was tested by enzyme linked immunosorbent assay, the mtROS level was detected by flow cytometry, and the immunofluorescence and co-localization of LC3 and mitochondria were observed under confocal laser scanning microscopy.
RESULTS:
Similar to the effect of Mito-TEMPO and contrary to the results of 3-MA treatment, TRQ could significantly reduce the expressions of NLRP3, Caspase-1 p20, and autophagy adaptor P62, promote the expression of autophagy marker LC3II, enhance the mitochondrial fluorescence intensity, and inhibit the release of mtROS and IL-1β (all P<0.01). Moreover, LC3 was co-localized with mitochondria, confirming the type of mitophagy.
CONCLUSION
TRQ could reduce the level of mtROS by promoting mitophagy in macrophages infected with influenza A virus, thus inhibiting the activation of NLRP3 inflammasome and the release of IL-1β, and attenuating the inflammatory response.
Mitophagy/drug effects*
;
NLR Family, Pyrin Domain-Containing 3 Protein/metabolism*
;
Animals
;
Macrophages/virology*
;
Inflammasomes/drug effects*
;
Drugs, Chinese Herbal/pharmacology*
;
Mice
;
Mitochondria/metabolism*
;
Reactive Oxygen Species/metabolism*
;
Influenza A virus/physiology*
;
Interleukin-1beta/metabolism*
;
Cell Line
;
Injections
9.Quercetin Confers Protection against Sepsis-Related Acute Respiratory Distress Syndrome by Suppressing ROS/p38 MAPK Pathway.
Wei-Chao DING ; Juan CHEN ; Quan LI ; Yi REN ; Meng-Meng WANG ; Wei ZHANG ; Xiao-Hang JI ; Xin-Yao WU ; Shi-Nan NIE ; Chang-Bao HUANG ; Zhao-Rui SUN
Chinese journal of integrative medicine 2025;31(11):1011-1020
OBJECTIVE:
To identify the underlying mechanism by which quercetin (Que) alleviates sepsis-related acute respiratory distress syndrome (ARDS).
METHODS:
In vivo, C57BL/6 mice were assigned to sham, cecal ligation and puncture (CLP), and CLP+Que (50 mg/kg) groups (n=15 per group) by using a random number table. The sepsisrelated ARDS mouse model was established using the CLP method. In vitro, the murine alveolar macrophages (MH-S) cells were classified into control, lipopolysaccharide (LPS), LPS+Que (10 μmol/L), and LPS+Que+acetylcysteine (NAC, 5 mmol/L) groups. The effect of Que on oxidative stress, inflammation, and apoptosis in mice lungs and MH-S cells was determined, and the mechanism with reactive oxygen species (ROS)/p38 mitogen-activated protein kinase (MAPK) pathway was also explored both in vivo and in vitro.
RESULTS:
Que alleviated lung injury in mice, as reflected by a reversal of pulmonary histopathologic changes as well as a reduction in lung wet/dry weight ratio and neutrophil infiltration (P<0.05 or P<0.01). Additionally, Que improved the survival rate and relieved gas exchange impairment in mice (P<0.01). Que treatment also remarkedly reduced malondialdehyde formation, superoxide dismutase and catalase depletion, and cell apoptosis both in vivo and in vitro (P<0.05 or P<0.01). Moreover, Que treatment diminished the release of inflammatory factors interleukin (IL)-1β, tumor necrosis factor-α, and IL-6 both in vivo and in vitro (P<0.05 or P<0.01). Mechanistic investigation clarifified that Que administration led to a decline in the phosphorylation of p38 MAPK in addition to the suppression of ROS expression (P<0.01). Furthermore, in LPS-induced MH-S cells, ROS inhibitor NAC further inhibited ROS/p38 MAPK pathway, as well as oxidative stress, inflammation, and cell apoptosis on the basis of Que treatment (P<0.05 or P<0.01).
CONCLUSION
Que was found to exert anti-oxidative, anti-inflammatory, and anti-apoptotic effects by suppressing the ROS/p38 MAPK pathway, thereby conferring protection for mice against sepsis-related ARDS.
Animals
;
Sepsis/drug therapy*
;
Quercetin/therapeutic use*
;
Respiratory Distress Syndrome/enzymology*
;
p38 Mitogen-Activated Protein Kinases/metabolism*
;
Mice, Inbred C57BL
;
Reactive Oxygen Species/metabolism*
;
Apoptosis/drug effects*
;
Male
;
Oxidative Stress/drug effects*
;
MAP Kinase Signaling System/drug effects*
;
Lung/drug effects*
;
Mice
;
Lipopolysaccharides
;
Macrophages, Alveolar/pathology*
;
Inflammation/pathology*
;
Protective Agents/therapeutic use*
10.High glucose induces pro-inflammatory polarization of macrophages by inhibiting immune-responsive gene 1 expression.
Wei LUO ; Yuhang WANG ; Yansong LIU ; Yuanyuan WANG ; Lei AI
Journal of Southern Medical University 2025;45(1):1-9
OBJECTIVES:
To investigate the effect of high glucose on macrophage polarization and the role of immune-responsive gene 1 (IRG1) in mediating its effect.
METHODS:
RAW264.7 cells were transfected with IRG1-overexpressing plasmid or IRG1 siRNA via electroporation and cultured in either normal or high glucose for 72 h to observe the changes in cell viability and morphology using CCK-8 assay and phase contrast microscopy. The protein levels of IRG1, iNOS, Arg-1, IL-1β and IL-10 in the treated cells were detected with Western blotting, and the fluorescence intensities of iNOS and Arg-1 were detected using immunofluorescence assay. The protein levels of IL-1β and IL-10 in the culture medium were determined with ELISA.
RESULTS:
High glucose exposure significantly reduced IRG1 and Arg-1 expressions, increased iNOS and IL-1β expressions and IL-1β secretion, and decreased IL-10 level in RAW264.7 cells. Transfection with the IRG1-overexpressing plasmid provided the cells with obvious resistance to high glucose-induced changes in iNOS, Arg-1, IL-1β and IL-10, whereas IRG1 knockdown further enhanced the effects of high glucose exposure on Arg-1 expression and the expression and secretion of IL-10.
CONCLUSIONS
High glucose promotes M1 polarization of the macrophages possibly through a mechanism to inhibit the expression of IRG1 protein, thus leading to chronic inflammatory response.
Animals
;
Mice
;
Macrophages/drug effects*
;
Glucose/pharmacology*
;
Interleukin-10/metabolism*
;
Nitric Oxide Synthase Type II/metabolism*
;
RAW 264.7 Cells
;
Interleukin-1beta/metabolism*
;
Arginase/metabolism*
;
RNA, Small Interfering/genetics*
;
Transfection
;
Inflammation

Result Analysis
Print
Save
E-mail