1.Nano-vesicles derived from M1 macrophages affect the development of mouse endometriosis by polarizing M2 macrophages to M1 phenotype.
Qiu MENG ; Xingrun XIE ; Shouguo HUANG
Chinese Journal of Cellular and Molecular Immunology 2023;39(9):807-815
Objective To explore whether nano-vesicles derived from M1 macrophages (M1-NVs) can reprogram M2 macrophages into M1 phenotype and further affect the development of endometriosis (EMS). Methods Extracellular vesicles (EVs) were isolated from macrophage culture supernatant by differential centrifugation. Immunofluorescence cytochemistry was used to detect the expression of vimentin, CD31 and F4/80 to identify endometrial stromal cells (EMS-ESCs), HUVECs and polarized peritoneal macrophages of EMS patients. M1-NVs were prepared by filtering cell suspension through (5, 1, 0.4, 0.22)μm polycarbonate membrane filters after syringe aspiration at 0-4 DegreesCelsius. Flow cytometry was used to analyze the polarization of RAW264.7 mouse peritoneal macrophages in vitro, and reverse transcription PCR (RT-qPCR) was employed to detect mRNA expression of VEGF, CD86, interleukin-6 (IL-6), IL-1β, tumor necrosis factor α (TNF-α), arginase 1 (Arg1), CD163, CD206, and IL-10. PKH67-labeled M1-NVs were co-cultured with EMS-ESCs, HUVECs and macrophages. And tubule formation experiments were conducted to assess the impact of M1-NVs on the tubule formation of HUVECs. TranswellTM invasion and migration assays were employed to evaluate changes in the migration and invasion abilities of EMS-ESCs. Results By monitoring the contents of NVs, it was found that NVs contained much more protein and other bioactive particles than the same amount of EVs; immunofluorescence staining results showed that PKH67 labeled M1-NVs were internalized by EMS-ESCs, HUVECs and macrophages when co-cultured. The results of flow cytometry and RT-qPCR multi-target analysis showed that after treatment with different concentrations of M1-NVs or M0-NVs, 20 μg/mL of M1-NVs could effectively reprogram M2 macrophages into M1 macrophages compared with M0-NVs. TransewellTM results showed that compared with the blank group and M0-NVs group, the number of EMS-ESCs migrating from the upper chamber to the lower chamber after M1-NV treatment was significantly reduced, while the number of EMS-ESCs treated with M2NVs increased significantly. The invasion situation was similar to the migration situation, indicating that M1-NVs directly or indirectly inhibited invasion, migration and tubule formation of EMS-ESCs in vitro. Conclusion M1-NVs reprogrammes M2 macrophages into M1 macrophages by internalization of primary cells and macrophages, thereby inhibiting invasion, migration and angiogenesis of EMS-ESCs, and further hindering the occurrence and development of EMS.
Female
;
Humans
;
Animals
;
Mice
;
Endometriosis
;
Macrophages
;
Macrophages, Peritoneal
;
Coculture Techniques
2.Estradiol inhibits differentiation of mouse macrophage into a pro-inflammatory phenotype by upregulating the IRE1α-XBP1 signaling axis.
Ling Jian ZHUO ; Shuo Chen WANG ; Xing LIU ; Bao An CHEN ; Xiang LI
Journal of Southern Medical University 2022;42(3):432-437
OBJECTIVE:
To explore the mechanism by which estradiol modulates the immunophenotype of macrophages through the endoplasmic reticulum stress pathway.
METHODS:
Peritoneal macrophages isolated from C57 mice were cultured in the presence of 60 ng/mL interferon-γ (IFN-γ) followed by treatment with estradiol (1.0 nmol/L) alone, estradiol with estrogen receptor antagonist (Acolbifene, 4 nmol/L), estradiol with IRE1α inhibitor (4 μ 8 C), or estradiol with IRE1α agonist. After the treatments, the expression levels of MHC-Ⅱ, iNOS and endoplasmic reticulum stress marker proteins IRE1α, eIF2α and ATF6 in the macrophages were detected with Western blotting, and the mRNA levels of TGF-β, IL-6, IL-10 and TNF-α were detected with RT-PCR.
RESULTS:
Estrogen treatment of the macrophages significantly decreased the expressions of M1-related proteins MHC-Ⅱ (P=0.021) and iNOS (P < 0.001) and the mRNA expressions of TNF-α (P=0.003) and IL-6 (P=0.004), increased the mRNA expression of TGF-β (P=0.002) and IL-10 (P=0.008), and up-regulated the protein expressions of IRE1α (P < 0.001) and its downstream transcription factor XBP-1 (P < 0.001). Addition of the estrogen inhibitor obviously blocked the effect of estrogen. Compared with estrogen treatment alone, combined treatment of the macrophages with estrogen and the IRE1α inhibitor 4 μ 8 C significantly up-regulated the protein expressions of MHC-Ⅱ (P=0.002) and iNOS (P=0.003) and the mRNA expressions of TNF-α (P=0.003) and IL-6 (P=0.024), and obviously down-regulated the mRNA expression of TGF-β (P < 0.001) and IL-10 (P < 0.001); these changes were not observed in cells treated with estrogen and the IRE1α agonist.
CONCLUSION
Estrogen can inhibit the differentiation of murine macrophages into a pro-inflammatory phenotype by up-regulating the IRE1α-XBP-1 signaling axis, thereby producing an inhibitory effect on inflammatory response.
Animals
;
Cell Differentiation/drug effects*
;
Endoribonucleases/metabolism*
;
Estradiol/pharmacology*
;
Estrogens/metabolism*
;
Interleukin-10
;
Interleukin-6/metabolism*
;
Macrophages, Peritoneal/metabolism*
;
Mice
;
Phenotype
;
Protein Serine-Threonine Kinases/metabolism*
;
RNA, Messenger/metabolism*
;
Signal Transduction/drug effects*
;
Transforming Growth Factor beta/metabolism*
;
Tumor Necrosis Factor-alpha/metabolism*
;
Up-Regulation/drug effects*
;
X-Box Binding Protein 1/metabolism*
3.Characterization of specialized innate immune cells in the omentum.
Acta Physiologica Sinica 2021;73(2):175-180
The great omentum is an intraperitoneal organ and plays an important role in protecting the environment of the peritoneal cavity. Several specialized innate immune cells including B1 cells and resident macrophages are found in the omentum, which may be attributed to the unique niche and its special stromal cells. However, it is not clear how these omental innate immune cells contribute to the peritoneal immunity. This review attempts to summarize the latest research on the omental innate immunity and discuss its involvement in the immune response of the peritoneal cavity.
Immunity, Innate
;
Macrophages
;
Omentum
;
Peritoneal Cavity
;
Stromal Cells
4.Rhodanthpyrone A and B play an anti-inflammatory role by suppressing the nuclear factor-κB pathway in macrophages
Kyeong Su KIM ; Chang Yeob HAN ; Young Taek HAN ; Eun Ju BAE
The Korean Journal of Physiology and Pharmacology 2019;23(6):493-499
Macrophage-associated inflammation is crucial for the pathogenesis of diverse diseases including metabolic disorders. Rhodanthpyrone (Rho) is an active component of Gentiana rhodantha, which has been used in traditional Chinese medicine to treat inflammation. Although synthesis procedures of RhoA and RhoB were reported, the biological effects of the specific compounds have never been explored. In this study, the anti-inflammatory activity and mechanisms of action of RhoA and RhoB were studied in lipopolysaccharide (LPS)-stimulated macrophages. Pretreatment with RhoA and RhoB decreased inducible nitric oxide synthase and cyclooxygenase-2 expressions in RAW 264.7 cells and in thioglycollate-elicited mouse peritoneal macrophages. In addition, it downregulated transcript levels of several inflammatory genes in LPS-stimulated RAW 264.7 cells, including inflammatory cytokines/chemokines (Tnfa, Il6, and Ccl2) and inflammatory mediators (Nos2 and Ptgs2). Macrophage chemotaxis was also inhibited by treatment with the compounds. Mechanistic studies revealed that RhoA and RhoB suppressed the nuclear factor (NF)-κB pathway, but not the canonical mitogen activated protein kinase pathway, in LPS-stimulated condition. Moreover, the inhibitory effect of RhoA and RhoB on inflammatory gene expressions was attenuated by treatment with an NF-κB inhibitor. Our findings suggest that RhoA and RhoB play an anti-inflammatory role at least in part by suppressing the NF-κB pathway during macrophage-mediated inflammation.
Animals
;
Chemotaxis
;
Cyclooxygenase 2
;
Gene Expression
;
Gentiana
;
Inflammation
;
Interleukin-6
;
Macrophages
;
Macrophages, Peritoneal
;
Medicine, Chinese Traditional
;
Mice
;
Nitric Oxide Synthase Type II
;
Protein Kinases
;
RAW 264.7 Cells
5.Two Distinct Subsets Are Identified from the Peritoneal Myeloid Mononuclear Cells Expressing both CD11c and CD115
Moah SOHN ; Hye Young NA ; Seul Hye RYU ; Wanho CHOI ; Hyunju IN ; Hyun Soo SHIN ; Ji Soo PARK ; Dahee SHIM ; Sung Jae SHIN ; Chae Gyu PARK
Immune Network 2019;19(3):e15-
To this date, the criteria to distinguish peritoneal macrophages and dendritic cells (DCs) are not clear. Here we delineate the subsets of myeloid mononuclear cells in the mouse peritoneal cavity. Considering phenotypical, functional, and ontogenic features, peritoneal myeloid mononuclear cells are divided into 5 subsets: large peritoneal macrophages (LPMs), small peritoneal macrophages (SPMs), DCs, and 2 MHCII⁺CD11c⁺CD115⁺ subpopulations (i.e., MHCII⁺CD11c⁺CD115⁺CD14⁻CD206⁻ and MHCII⁺CD11c⁺CD115⁺CD14⁺CD206⁺). Among them, 2 subsets of competent Ag presenting cells are demonstrated with distinct functional characteristics, one being DCs and the other being MHCII⁺CD11c⁺CD115⁺CD14⁻CD206⁻ cells. DCs are able to promote fully activated T cells and superior in expanding cytokine producing inflammatory T cells, whereas MHCII⁺CD11c⁺CD115⁺CD14⁻CD206⁻ cells generate partially activated T cells and possess a greater ability to induce Treg under TGF-β and retinoic acid conditions. While the development of DCs and MHCII⁺CD11c⁺CD115⁺CD14⁻CD206⁻ cells are responsive to the treatment of FLT3 ligand and GM-CSF, the number of LPMs, SPMs, and MHCII⁺CD11c⁺CD115⁺CD14⁺CD206⁺ cells are only influenced by the injection of GM-CSF. In addition, the analysis of gene expression profiles among MHCII⁺ peritoneal myeloid mononuclear cells reveals that MHCII⁺CD11c⁺CD115⁺CD14⁺CD206⁺ cells share high similarity with SPMs, whereas MHCII⁺CD11c⁺CD115⁺CD14⁻CD206⁻ cells are related to peritoneal DC2s. Collectively, our study identifies 2 distinct subpopulations of MHCII⁺CD11c⁺CD115⁺ cells, 1) MHCII⁺CD11c⁺CD115⁺CD14⁻CD206⁻ cells closely related to peritoneal DC2s and 2) MHCII⁺CD11c⁺CD115⁺CD14⁺CD206⁺ cells to SPMs.
Animals
;
Antigen Presentation
;
Dendritic Cells
;
Granulocyte-Macrophage Colony-Stimulating Factor
;
Macrophages
;
Macrophages, Peritoneal
;
Mice
;
Peritoneal Cavity
;
T-Lymphocytes
;
Transcriptome
;
Tretinoin
6.Perivascular Stem Cells Suppress Inflammasome Activation during Inflammatory Responses in Macrophages
Jeeyoung KIM ; Woo Jin KIM ; Kwon Soo HA ; Eun Taek HAN ; Won Sun PARK ; Se Ran YANG ; Seok Ho HONG
International Journal of Stem Cells 2019;12(3):419-429
BACKGROUND AND OBJECTIVES: Perivascular stem cells (PVCs) have been identified as precursors of mesenchymal stem cells (MSCs) that offer promising prospects for application in the development of cellular therapies. Although PVCs have been demonstrated to have greater therapeutic potential compared to bone marrow and adipose tissue-derived MSCs in various diseases, the regulatory role of PVCs on inflammasome activation during macrophage-mediated inflammatory responses has not been investigated.METHODS AND RESULTS: In this study, we found that the PVC secretome effectively alleviates secretion of both caspase-1 and interleukin-1β in lipopolysaccharide-primed and activated human and murine macrophages by blocking inflammasome activation and attenuating the production of mitochondrial reactive oxygen species (ROS). We further showed that the PVC secretome significantly reduces inflammatory responses and endoplasmic reticulum stress in peritoneal macrophages in a mouse model of monosodium urate-induced peritonitis. A cytokine antibody array analysis revealed that the PVC secretome contains high levels of serpin E1 and angiogenin, which may be responsible for the inhibitory effects on mitochondrial ROS generation as well as on inflammasome activation.CONCLUSIONS: Our results suggest that PVCs may be therapeutically useful for the treatment of macrophage- and inflammation-mediated diseases by paracrine action via the secretion of various biological factors.
Animals
;
Biological Factors
;
Bone Marrow
;
Endoplasmic Reticulum Stress
;
Humans
;
Inflammasomes
;
Inflammation
;
Macrophages
;
Macrophages, Peritoneal
;
Mesenchymal Stromal Cells
;
Mice
;
Peritonitis
;
Plasminogen Activator Inhibitor 1
;
Reactive Oxygen Species
;
Stem Cells
7.TLR/NF-κB independent signaling pathway in TNF-α suppression of diabetic MyD88-knockout mice after polysaccharides administration.
Tingting LIU ; Lingxiao WANG ; Xiaohui YANG ; Zhiqing YAO ; Huizhen CAI
Journal of Zhejiang University. Medical sciences 2018;47(1):35-40
OBJECTIVE:
: To investigate the effect of polysaccharides (LBPs) on TLR/NF-κB independent pathway and serum tumor necrosis factor (TNF-α) level in diabetic MyD88-knockout mice.
METHODS:
: Diabetes was induced by feeding high-fat/high-sugar diet and injection of low-dose streptozotocin in MyD88-knockout mice. The diabetic mice were randomly divided into model group, positive control group and LBPs group. The expressions of TRAM, TRIF, TRAF6, RIP1 and TNF-α mRNA and proteins in mouse peritoneal macrophages were detected by real-time RT-PCR and Western blotting after LBPs treatment for 3 month. Serum TNF-α was determined with ELISA kit.
RESULTS:
: Real time RT-PCR showed that compared with model group, the relative expressions of and mRNA in macrophages of LBPs group were significantly decreased and expression of was significantly increased (all <0.05). Expression of TRAM, TRIF, TRAF6, RIP1 and TNF-α proteins as well as serum TNF-α level had no significant difference between LBPs group and model group (all >0.05).
CONCLUSIONS
: LBPs may not inhibit serum TNF-α level through TLR/NF-κB independent pathway.
Animals
;
Diabetes Mellitus, Experimental
;
Drugs, Chinese Herbal
;
pharmacology
;
Gene Expression Regulation
;
drug effects
;
Macrophages, Peritoneal
;
drug effects
;
Mice
;
Mice, Knockout
;
Myeloid Differentiation Factor 88
;
genetics
;
NF-kappa B
;
genetics
;
Random Allocation
;
Signal Transduction
;
drug effects
;
Tumor Necrosis Factor-alpha
;
blood
;
metabolism
8.Immunological characteristics of peritoneal cavity and intra-abdominal infection.
Chinese Journal of Gastrointestinal Surgery 2018;21(12):1347-1350
Despite the evolution of aggressive surgical techniques, extensive methods of supportive care and a vast array of anti-microbial options, intra-abdominal infection (IAI) is still a challenging clinical issue. Especially, when progressed IAI with septic complications because of unbalanced immune responses, the prognosis will deteriorated significantly. Recent studies indicate that besides the natural immunological cells, including macrophages and neutrophils, local immunological characteristics of peritoneal cavity should be studied with great attention. Among them, the omentum is considered to be a visceral adipose tissue with unique immune function. The milky spots(MSs) formed by the accumulation of immune cells performs immune surveillance and has a lymph node-like immune function, which is very important for the immune defense of the abdominal cavity. B1 cells and two types of intrinsic lymphocytes(ILC2) in the peritoneal cavity, although belonging to the lymphatic lineage, may play an important role in abdominal infections, especially in the early stages of the disease, due to their rapid responsiveness and acquired immune function. Therefore, paying attention to the immunological characteristics of the peritoneal cavity, and elucidating the changes, functions and regulatory mechanisms of B1 cells and ILC2 around the MSs and their components in the process of IAI, in order to explore the immunomodulation targets of blocking the infection from local to systemic dissemination, may be the key to solving the clinical problem of severe IAI and improving prognosis.
Humans
;
Intraabdominal Infections
;
immunology
;
Lymphocytes
;
immunology
;
Macrophages
;
immunology
;
Omentum
;
immunology
;
Peritoneal Cavity
9.LPS-stimulated Macrophage Activation Affects Endothelial Dysfunction.
Naehwan BAEK ; Sohyun SIM ; Kyung Sun HEO
Journal of Bacteriology and Virology 2018;48(1):23-30
Intestinal microbiota is involved in the atherosclerotic process by development of an atheromatous core with foam cells in carotid arteries. It has reported that lipopolysaccharide (LPS) from Escherichia coli localizes in human atherosclerotic plaque and causes inflammation via interaction with toll like receptor 4. However, there is no evidence that whether LPS-activated macrophages regulate endothelial cell (EC) function. We evaluated whether LPS-activated macrophage acts as one of the stimulants activating EC and its underlying signaling pathways. Using Western blotting and quantitative reverse transcription-polymerase chain reaction (qRT-PCR), we confirmed that intraperitoneal injection with LPS increases iNOS protein and inflammatory cytokine, TNF-α and IL-6 mRNA expressions. To determine whether LPS-mediated macrophage inflammatory condition affects EC activation and inflammation, human umbilical vein endothelial cells (HUVECs) were incubated with isolated peritoneal macrophages from LPS-injected mice. Interestingly, p90RSK Serine 380 phosphorylation and protein expression were significantly increased by macrophage treatment in EC. Messenger RNA levels of vascular cell adhesion molecule 1 and p90RSK was increased, but endothelial nitric oxide synthase was decreased. In addition, NF-κB promoter activity, which plays an important role in the pathogenesis of inflammation, was strongly enhanced by the macrophage treatment in EC. We further evaluated the effects of LPS on EC function in the mouse aorta using en face staining. In agreement with in vitro result, p90RSK expression was strongly increased in the steady laminar flow region of the mouse aorta in mice injected with LPS. Together, our study demonstrates that p90RSK might be a one of the major therapeutic candidates for the prevention of vascular diseases mediated by LPS.
Animals
;
Aorta
;
Atherosclerosis
;
Blotting, Western
;
Carotid Arteries
;
Endothelial Cells
;
Escherichia coli
;
Foam Cells
;
Gastrointestinal Microbiome
;
Human Umbilical Vein Endothelial Cells
;
Humans
;
In Vitro Techniques
;
Inflammation
;
Injections, Intraperitoneal
;
Interleukin-6
;
Macrophage Activation*
;
Macrophages*
;
Macrophages, Peritoneal
;
Mice
;
Nitric Oxide Synthase Type III
;
Phosphorylation
;
Plaque, Atherosclerotic
;
RNA, Messenger
;
Serine
;
Toll-Like Receptor 4
;
Vascular Cell Adhesion Molecule-1
;
Vascular Diseases
10.Adenine attenuates lipopolysaccharide-induced inflammatory reactions.
Prashanta SILWAL ; Kyu LIM ; Jun Young HEO ; Jong IL PARK ; Uk NAMGUNG ; Seung Kiel PARK
The Korean Journal of Physiology and Pharmacology 2018;22(4):379-389
A nucleobase adenine is a fundamental component of nucleic acids and adenine nucleotides. Various biological roles of adenine have been discovered. It is not produced from degradation of adenine nucleotides in mammals but produced mainly during polyamine synthesis by dividing cells. Anti-inflammatory roles of adenine have been supported in IgE-mediated allergic reactions, immunological functions of lymphocytes and dextran sodium sulfate-induced colitis. However adenine effects on Toll-like receptor 4 (TLR4)-mediated inflammation by lipopolysaccharide (LPS), a cell wall component of Gram negative bacteria, is not examined. Here we investigated anti-inflammatory roles of adenine in LPS-stimulated immune cells, including a macrophage cell line RAW264.7 and bone marrow derived mast cells (BMMCs) and peritoneal cells in mice. In RAW264.7 cells stimulated with LPS, adenine inhibited production of pro-inflammatory cytokines TNF-α and IL-6 and inflammatory lipid mediators, prostaglandin E₂ and leukotriene B₄. Adenine impeded signaling pathways eliciting production of these inflammatory mediators. It suppressed IκB phosphorylation, nuclear translocation of nuclear factor κB (NF-κB), phosphorylation of Akt and mitogen activated protein kinases (MAPKs) JNK and ERK. Although adenine raised cellular AMP which could activate AMP-dependent protein kinase (AMPK), the enzyme activity was not enhanced. In BMMCs, adenine inhibited the LPS-induced production of TNF-α, IL-6 and IL-13 and also hindered phosphorylation of NF-κB and Akt. In peritoneal cavity, adenine suppressed the LPS-induced production of TNF-α and IL-6 by peritoneal cells in mice. These results show that adenine attenuates the LPS-induced inflammatory reactions.
Adenine Nucleotides
;
Adenine*
;
Animals
;
Bone Marrow
;
Cell Line
;
Cell Wall
;
Colitis
;
Cytokines
;
Dextrans
;
Gram-Negative Bacteria
;
Hypersensitivity
;
Inflammation
;
Interleukin-13
;
Interleukin-6
;
Lymphocytes
;
Macrophages
;
Mammals
;
Mast Cells
;
Mice
;
Mitogen-Activated Protein Kinases
;
Nucleic Acids
;
Peritoneal Cavity
;
Phosphorylation
;
Protein Kinases
;
Sodium
;
Toll-Like Receptor 4

Result Analysis
Print
Save
E-mail