Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Front Immunol ; 12: 680068, 2021.
Article in English | MEDLINE | ID: mdl-34025679

ABSTRACT

Toll-like receptors (TLRs) play critical roles in regulating the abnormal activation of the immune cells resulting in the pathogenesis of inflammation and autoimmune diseases. Pyruvate kinase M2 (PKM2), which governs the last step of glycolysis, is involved in multiple cellular processes and pathological conditions. However, little is known about the involvement of PKM2 in regulating TLR-mediated inflammation and autoimmunity. Herein, we investigated the role of PKM2 in the activation of the TLR pathways and the pathogenesis of inflammation and autoimmune diseases. The activation of TLR4, TLR7 and TLR9 pathways was found to induce the up-regulation of PKM2 expression in macrophages, dendritic cells (DCs) and B cells. The over-expression of PKM2 promotes the activation of TLR4, TLR7 and TLR9 pathways while interference with the PKM2 expression or the addition of the PKM2 inhibitor (PKM-IN) markedly inhibited the activation of TLR4, TLR7 and TLR9 pathways. Mechanistically, PKM2 augmented the activation of TLR4, TLR7 and TLR9 pathways by promoting the activation of the proline-rich tyrosine kinase 2 (Pyk2). Intriguingly, the PKM2 inhibitor PKM2-IN significantly protected the mice from the endotoxic shock mediated by the TLR4-agonist LPS. Additionally, it alleviated the progression in the TLR7-agonist imiquimod-mediated lupus mice and spontaneous lupus MRL/lpr mice. Moreover, PKM2 expression was highly elevated in the monocytes, DCs and B cells from systemic lupus erythematous (SLE) patients compared with those from the healthy donors. Besides, the PKM2 expression level was positively correlated with the degree of activation of these immune cells. In summary, PKM2 contributed to TLR-mediated inflammation and autoimmunity and can be a valuable target to control inflammation and autoimmunity.


Subject(s)
Autoimmunity , Carrier Proteins/metabolism , Focal Adhesion Kinase 2/metabolism , Inflammation/etiology , Inflammation/metabolism , Membrane Proteins/metabolism , Thyroid Hormones/metabolism , Toll-Like Receptors/metabolism , Animals , Autoimmune Diseases/etiology , Autoimmune Diseases/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Biomarkers , Carrier Proteins/antagonists & inhibitors , Cell Survival , Disease Models, Animal , Disease Susceptibility , Female , Inflammation/diagnosis , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/metabolism , Lupus Erythematosus, Systemic/pathology , Lymphocyte Activation/immunology , Macrophages/immunology , Macrophages/metabolism , Membrane Proteins/antagonists & inhibitors , Mice , Mice, Inbred MRL lpr , Models, Biological , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , Toll-Like Receptor 7/metabolism , Toll-Like Receptor 9/metabolism , Thyroid Hormone-Binding Proteins
2.
Biochim Biophys Acta Mol Basis Dis ; 1866(1): 165554, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31513833

ABSTRACT

Activation of interferon (IFN)-I signaling in B cells contributes to the pathogenesis of systemic lupus erythematosus (SLE). Recent studies have shown that myeloid-derived suppressor cells (MDSCs) significantly expand in SLE patients and lupus-prone MRL/lpr mice and contribute to the pathogenesis of SLE. However, the role of SLE-derived MDSCs in regulating IFN-I signaling activation of B cells remains unknown. Here, we demonstrate that expansions of MDSCs, including granulocyte (G)-MDSCs and monocytic (M)-MDSCs, during the progression of SLE were correlated with the IFN-I signature of B cells. Interestingly, G-MDSCs from MRL/lpr mice, but not M-MDSCs, could significantly promote IFN-I signaling activation of B cells and contribute to the pathogenesis of SLE. Mechanistically, we identified that the long non-coding RNA NEAT1 was over-expressed in G-MDSCs from MRL/lpr mice and could induce the promotion of G-MDSCs on IFN-I signaling activation of B cells through B cell-activating factor (BAFF) secretion. Importantly, NEAT1 deficiency significantly attenuated the lupus symptoms in pristane-induced lupus mice. In addition, there was a positive correlation between NEAT1 and BAFF with the IFN signature in SLE patients. In conclusion, G-MDSCs may contribute to the IFN signature in SLE B cells through the NEAT1-BAFF axis, highlighting G-MDSCs as a potential therapeutic target to treat SLE.


Subject(s)
B-Cell Activating Factor/metabolism , B-Lymphocytes/metabolism , Interferon Type I/metabolism , Lupus Erythematosus, Systemic/metabolism , Myeloid-Derived Suppressor Cells/metabolism , RNA, Long Noncoding/metabolism , Signal Transduction/physiology , Animals , Cytokines/metabolism , Disease Progression , Female , Granulocytes/metabolism , Granulocytes/pathology , Humans , Kidney/metabolism , Kidney/pathology , Lupus Erythematosus, Systemic/pathology , Mice , Mice, Inbred C57BL , Mice, Inbred MRL lpr , Myeloid-Derived Suppressor Cells/pathology
3.
Am J Transl Res ; 11(5): 3029-3038, 2019.
Article in English | MEDLINE | ID: mdl-31217872

ABSTRACT

Immune-mediated liver injury plays a crucial role in the pathogenesis of liver diseases, which can result from viral infections, autoimmunity, alcohol intake, and drug use. Concanavalin A (Con A)-induced hepatitis is a well-characterized murine model with similar pathophysiology to that of human viral and autoimmune hepatitis. Capsaicin, a selective agonist of the transient potential vanilloid subfamily member 1 (TRPV1) receptor, exhibits anti-inflammatory effects on various causes of inflammation. In the present study, we investigated the effect of capsaicin on Con A-induced hepatitis. Capsaicin (1 mg/kg body weight) was administered by intraperitoneal injection, after which (30 minutes), the mice were challenged intravenously with Con A (20 µg/g body weight). We collected serum for plasma transaminase analysis. Pro-inflammatory cytokine levels and hepatocyte apoptosis were assayed by ELISA and TUNEL, respectively. Liver samples were collected for real-time PCR, hematoxylin and eosin staining, and measuring oxidative stress and myeloperoxidase levels. Activation of splenocytes and hepatic mononuclear cells was analyzed by flow cytometry. Compared with control, the capsaicin-treated group showed significantly decreased aminotransferase levels and markedly prolonged mouse survival. Capsaicin pretreatment also attenuated hepatocyte apoptosis and oxidative stress. Furthermore, tumor necrosis factor-α and interferon-γ levels in serum and liver were significantly suppressed, while the percentage of myeloid-derived suppressor cells increased after capsaicin pretreatment. Our findings indicate that capsaicin pretreatment protects mice from Con A-induced hepatic damage and is partially involved in inhibiting hepatocyte apoptosis, oxidative stress, and inflammatory mediators as well as regulating activation and recruitment of intrahepatic leukocytes.

4.
Front Immunol ; 10: 215, 2019.
Article in English | MEDLINE | ID: mdl-30809230

ABSTRACT

Dysregulation of macrophage has been demonstrated to contribute to aberrant immune responses and inflammatory diseases. CD11b, expressed on macrophages, plays a critical role in regulating pathogen recognition, phagocytosis, and cell survival. In the present study, we explored the effect of leukadherin-1 (LA1), an agonist of CD11b, on regulating LPS-induced pro-inflammatory response in macrophages and endotoxic shock. Intriguingly, we found that LA1 could significantly reduce mortalities of mice and alleviated pathological injury of liver and lung in endotoxic shock. In vivo studies showed that LA1-induced activation of CD11b significantly inhibited the LPS-induced pro-inflammatory response in macrophages of mice. Moreover, LA1-induced activation of CD11b significantly inhibited LPS/IFN-γ-induced pro-inflammatory response in macrophages by inhibiting MAPKs and NF-κB signaling pathways in vitro. Furthermore, the mice injected with LA1-treated BMDMs showed fewer pathological lesions than those injected with vehicle-treated BMDMs in endotoxic shock. In addition, we found that activation of TLR4 by LPS could endocytose CD11b and activation of CD11b by LA1 could endocytose TLR4 in vitro and in vivo, subsequently blocking the binding of LPS with TLR4. Based on these findings, we concluded that LA1-induced activation of CD11b negatively regulates LPS-induced pro-inflammatory response in macrophages and subsequently protects mice from endotoxin shock by partially blocking LPS-TLR4 interaction. Our study provides a new insight into the role of CD11b in the pathogenesis of inflammatory diseases.


Subject(s)
Antigens, CD1/metabolism , Macrophage Activation/immunology , Macrophages/immunology , Macrophages/metabolism , Shock, Septic/etiology , Shock, Septic/metabolism , Toll-Like Receptor 4/metabolism , Animals , Benzoates/pharmacology , Biomarkers , Dendritic Cells/immunology , Dendritic Cells/metabolism , Endocytosis , Female , Kaplan-Meier Estimate , Lipopolysaccharides/immunology , Liver Diseases/etiology , Liver Diseases/pathology , Macrophage Activation/drug effects , Mice , Models, Biological , Mortality , Shock, Septic/complications , Shock, Septic/mortality , Thiohydantoins/pharmacology
5.
Biochim Biophys Acta Mol Basis Dis ; 1865(3): 535-546, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30557700

ABSTRACT

Myeloid-derived suppressor cells (MDSCs) play an immunosuppressive role in the pathogenesis of inflammatory diseases. CD180, a TLR-like protein, can regulate the proliferation and activation of immune cells. However, the roles of CD180 in regulating the accumulation and function of MDSCs have not been investigated. Here, we found that, compared with non-treated controls, the expression of CD180 was significantly elevated in MDSCs, especially granulocytic MDSCs (G-MDSCs), from mice challenged with lipopolysaccharide (LPS). Ligation of CD180 by the anti-CD180 antibody not only blocked the expansion of MDSCs by preventing the phosphorylation of signal transducer and activator of transcription 3 (STAT3), but also reduced the immunosuppressive activity of MDSCs on M1 macrophage polarization through inhibition of Arg-1 expression in vitro. In vivo studies showed that injection of anti-CD180 antibody significantly aggravated pathological lesions in mice challenged with LPS. Furthermore, injection of anti-CD180 antibody inhibited the accumulation of G-MDSCs in mice challenged with LPS and reduced the immunosuppressive activity of G-MDSCs on M1 macrophage polarization. Based on these findings, we conclude that ligation of CD180 contributes to the pathogenesis of endotoxic shock by inhibiting the accumulation and immunosuppressive activity of G-MDSCs, thus providing insight into the function of CD180 in inflammatory diseases.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antigens, CD/immunology , Myeloid-Derived Suppressor Cells/immunology , STAT3 Transcription Factor/physiology , Shock, Septic/chemically induced , Shock, Septic/immunology , Animals , Cell Proliferation/drug effects , Cells, Cultured , Female , Inflammation/chemically induced , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Myeloid-Derived Suppressor Cells/cytology , Myeloid-Derived Suppressor Cells/drug effects , Protein Binding , Shock, Septic/metabolism , Signal Transduction/drug effects , Signal Transduction/immunology
6.
Inflammation ; 41(6): 2090-2100, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30143931

ABSTRACT

Endotoxin shock is a life-threatening response caused by a disordered immune response to an infection. MDSCs are accumulated and play a protective role in the pathogenesis of endotoxin shock. However, the regulation of MDSCs by small molecule remains unrevealed. Here, we report that arctigenin, a small molecule extracted from Arctium lappa, induces accumulation of functional MDSCs. Arctigenin was able to ameliorate LPS-induced inflammation through accumulating MDSCs, especially granulocytic MDSCs (G-MDSCs), and enhancing the immunosuppressive function of MDSCs in vivo and in vitro. Mechanistically, arctigenin promoted the accumulation of MDSCs through upregulating miR-127-5p which targets the 3'UTR of interferon regulatory factor-8 (IRF8) mRNA. In addition, arctigenin enhanced the immunosuppressive activity of MDSCs on M1 macrophage polarization by elevating the expression of arginase 1 (Arg-1) and inducible nitric oxide synthase (iNOS). Our study provides new insights into the regulation of functional MDSCs by arctigenin in exerting immune responses and pathogenesis of inflammatory diseases.


Subject(s)
Furans/pharmacology , Inflammation/prevention & control , Lignans/pharmacology , Myeloid-Derived Suppressor Cells/immunology , Shock, Septic/pathology , Animals , Arginase/metabolism , Furans/therapeutic use , Interferon Regulatory Factors/genetics , Lignans/therapeutic use , Lipopolysaccharides , Mice , MicroRNAs/drug effects , Myeloid-Derived Suppressor Cells/drug effects , Nitric Oxide Synthase Type II/metabolism , RNA, Messenger , Shock, Septic/metabolism
7.
Biochim Biophys Acta Mol Basis Dis ; 1863(11): 2796-2807, 2017 11.
Article in English | MEDLINE | ID: mdl-28802852

ABSTRACT

Autophagy extensively participates in immune responses and inflammatory diseases. Myeloid-derived suppressor cells (MDSCs) are derived from CD11b+Gr1+ cells under pathological conditions and play an immunosuppressive role in the pathogenesis of cancer and inflammatory diseases. However, the role of autophagy in regulating the accumulation and activity of MDSCs remains unknown. In the present study, we evaluated the effects and mechanisms of autophagy on regulating accumulation and activity of MDSCs. We first found that granulocytic MDSCs (G-MDSCs), but not monocytic MDSCs (M-MDSCs), were accumulated in mice challenged by lipopolysaccharide (LPS) and showed an elevated autophagy activity. Pharmacological inhibition of autophagy significantly enhanced accumulation of G-MDSCs in vivo and in vitro. Notably, inhibition of autophagy enhanced the immunosuppressive activity of G-MDSCs on M1 macrophage polarization by promoting reactive oxygen species (ROS) production. Inhibition of autophagy promotes the phosphorylation of signal transducer and activator of transcription 3 (STAT3) in G-MDSCs, which is required for the accumulation and activity of MDSCs. In addition, in vivo pharmacological inhibition of autophagy significantly attenuated the condition of mice challenged by LPS. Thus, we conclude that inhibition of autophagy contributes to accumulation and immunosuppressive function of G-MDSCs by promoting the activation of STAT3 signaling, suggesting that autophagy may play a critical role in regulating accumulation and activity of MDSCs. Our study provides new insights into understanding the mechanisms of autophagy in regulating immune responses and pathogenesis of inflammatory diseases.


Subject(s)
Autophagy/immunology , Granulocytes/immunology , Myeloid-Derived Suppressor Cells/immunology , STAT3 Transcription Factor/immunology , Shock, Septic/immunology , Signal Transduction/immunology , Animals , Autophagy/drug effects , Granulocytes/pathology , Lipopolysaccharides/toxicity , Mice , Myeloid-Derived Suppressor Cells/pathology , Shock, Septic/chemically induced , Shock, Septic/pathology , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...