Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Br J Cancer ; 130(9): 1542-1551, 2024 May.
Article in English | MEDLINE | ID: mdl-38461171

ABSTRACT

BACKGROUND: Tumor cells continue to evolve the metastatic potential in response to signals provided by the external microenvironment during metastasis. Platelets closely interact with tumor cells during hematogenous metastasis and facilitate tumor development. However, the molecular mechanisms underlying this process are not fully understood. METHODS: RNA-sequencing was performed to screen differentially expressed genes mediated by platelets. The effects of platelet and CD39 on tumor metastasis were determined by experimental metastasis models with WT, NCG and CD39-/- mice. RESULTS: RNA-sequencing results showed that platelets significantly up-regulated CD39 expression in tumor cells. CD39 is a novel immune checkpoint molecule and a key driver of immunosuppression. Our data provided evidence that the expression of CD39 was enhanced by platelets in a platelet-tumor cell contact dependent manner. Although the role of CD39 expressed by immune cells is well established, the effect of CD39 expressed by tumor cells on tumor cell behavior, anti-tumor immunity and tumor metastasis is unclear. We found that CD39 promoted tumor cell invasion, but had no effect on proliferation and migration. Notably, we showed that the ability of platelets to prime tumor cells for metastasis depends on CD39 in the experimental tumor metastasis model. CD39 silencing resulted in fewer experimental metastasis formation, and this anti-metastasis effect was significantly reduced in platelet-depleted mice. Furthermore, overexpression of CD39 in tumor cells promoted metastasis. In order to eliminate the effect of CD39 expressed in cells other than tumor cells, we detected tumor metastasis in CD39-/- mice and obtained similar results. Moreover, overexpression of CD39 in tumor cells inhibited antitumor immunity. Finally, the data from human samples also supported our findings. CONCLUSIONS: Our study shows that direct contact with platelets induces CD39 expression in tumor cells, leading to immune suppression and promotion of metastasis.


Subject(s)
Antigens, CD , Apyrase , Blood Platelets , Neoplasm Metastasis , Animals , Apyrase/genetics , Apyrase/metabolism , Blood Platelets/metabolism , Blood Platelets/pathology , Mice , Antigens, CD/genetics , Antigens, CD/metabolism , Humans , Cell Line, Tumor , Female , Mice, Knockout , Cell Movement , Tumor Microenvironment/immunology , Gene Expression Regulation, Neoplastic
2.
Inflammation ; 46(1): 418-431, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36171490

ABSTRACT

Endotoxin shock remains one of the major causes of mortality worldwide. Pyruvate dehydrogenase kinase (PDK) 2 is an important regulatory enzyme involved in glucose metabolism. The purpose of this study was to determine the regulatory effect of PDK2 on LPS-induced endotoxin shock and explore the mechanisms in vivo and in vitro. Here, we showed that PDK2 contributed to Toll-like receptor (TLR)-mediated inflammation. Lipopolysaccharide (LPS) activation of TLR4 pathways resulted in PDK2 upregulation in macrophages and dendritic cells (DCs). PDK2 overexpression enhanced TLR4 signaling pathway activation, whereas downregulating PDK2 expression inhibited TLR4 signaling pathway activation. Pharmacological inhibition of PDK2 significantly decreased the mortality rate and alleviated pathological injury in the lungs and livers of LPS-challenged mice, while significantly suppressing proinflammatory cytokine production. Thus, we confirmed that PDK2 is involved in LPS-induced endotoxin shock by modulating TLR4-mitogen-activated protein kinase signaling and inducing the production of proinflammatory cytokines in macrophages and DCs. Our findings highlight the importance of PDK2 as a novel target to treat septic shock.


Subject(s)
Mitogen-Activated Protein Kinases , Shock, Septic , Animals , Mice , Lipopolysaccharides/pharmacology , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , Shock, Septic/metabolism , Toll-Like Receptor 4/metabolism
3.
Front Immunol ; 13: 960852, 2022.
Article in English | MEDLINE | ID: mdl-36032104

ABSTRACT

In recent studies, NKG2A is revealed to be a key immune checkpoint for both natural killer (NK) cells and CD8+ T cells. It form heterodimer receptors with CD94, and targets the peptide-presenting human leukocyte antigen-E (HLA-E) molecules. Upon crosslinking, NKG2A/CD94 delivers inhibitory signals for NK cells and CD8+ T cells, while blocking NKG2A can effectively unleash functions of these cytotoxic lymphocytes. The interaction between NKG2A and HLA-E contributes to tumor immune escape, and NKG2A-mediated mechanisms are currently being exploited to develop potential antitumor therapeutic strategies. In addition, growing evidence shows that NKG2A also plays important roles in other immune-related diseases including viral infections, autoimmune diseases, inflammatory diseases, parasite infections and transplant rejection. Therefore, the current work focuses on describing the effect of NKG2A on immune regulation and exploring its potential role in immune-mediated disorders.


Subject(s)
Immunity , NK Cell Lectin-Like Receptor Subfamily C , CD8-Positive T-Lymphocytes , HLA Antigens , Histocompatibility Antigens Class I , Humans , Killer Cells, Natural , NK Cell Lectin-Like Receptor Subfamily C/immunology
4.
Int Immunopharmacol ; 109: 108799, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35525232

ABSTRACT

The type III interferon family (IFN-III), including IFN-λ3 [interleukin (IL)-28B], has antiviral, anti-tumor, and immunomodulatory activities. Although the IL-28B anti-tumor effect has been extensively explored, its underlying mechanism remains unclear. Here, we explored IL-28B effects on colon cancer. Our results show that IL-28B significantly inhibits colon cancer progression in a mouse MC38 tumor cell colonization model and colitis-associated colorectal tumor model. Interestingly, IL-28B does not directly promote apoptosis or inhibit MC38 tumor cell proliferation in vitro. Rather, IL-28B treatment has indirect anti-tumor activity by downregulating tumor-associated macrophages. Furthermore, IL-28B inhibits M2 macrophage polarization in vitro, while also halting M2 macrophage differentiation predominantly via inhibition of the signal transducer and activator of transcription (STAT)3 and c-Jun N-terminal kinase (JNK) signaling pathways. Our findings revealed that IL-28B inhibits M2 macrophages in the tumor microenvironment to delay colon cancer progression. These findings provide novel evidence of IL-28B anti-tumor and immunomodulatory activities.


Subject(s)
Colonic Neoplasms , Tumor-Associated Macrophages , Animals , Antiviral Agents/pharmacology , Cell Line, Tumor , Colonic Neoplasms/metabolism , Macrophages , Mice , Signal Transduction , Tumor Microenvironment
5.
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
6.
Front Immunol ; 12: 654960, 2021.
Article in English | MEDLINE | ID: mdl-33859648

ABSTRACT

B and T lymphocyte attenuator (BTLA) is one of the most important cosignaling molecules. It belongs to the CD28 superfamily and is similar to programmed cell death-1 (PD-1) and cytotoxic T lymphocyte associated antigen-4 (CTLA-4) in terms of its structure and function. BTLA can be detected in most lymphocytes and induces immunosuppression by inhibiting B and T cell activation and proliferation. The BTLA ligand, herpesvirus entry mediator (HVEM), does not belong to the classic B7 family. Instead, it is a member of the tumor necrosis factor receptor (TNFR) superfamily. The association of BTLA with HVEM directly bridges the CD28 and TNFR families and mediates broad and powerful immune effects. Recently, a large number of studies have found that BTLA participates in numerous physiopathological processes, such as tumor, inflammatory diseases, autoimmune diseases, infectious diseases, and transplantation rejection. Therefore, the present work aimed to review the existing knowledge about BTLA in immunity and summarize the diverse functions of BTLA in various immune disorders.


Subject(s)
Immune System Diseases/etiology , Immune System Diseases/metabolism , Immunity , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Animals , Autoimmunity , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Disease Susceptibility , Graft Rejection/etiology , Graft Rejection/metabolism , Graft vs Host Disease/etiology , Graft vs Host Disease/metabolism , Humans , Organ Transplantation/adverse effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
7.
Inflammation ; 44(2): 671-681, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33083887

ABSTRACT

Fulminant hepatitis (FH) is an acute clinical disease with a poor prognosis and high mortality rate. The purpose of this study was to determine the protective effect of the Toll-like receptor 4 (TLR4) inhibitor TAK-242 on lipopolysaccharide (LPS)/D-galactosamine (D-GalN)-induced explosive hepatitis and explore in vivo and in vitro mechanisms. Mice were pretreated with TAK-242 for 3 h prior to LPS (10 µg/kg)/D-GalN (250 mg/kg) administration. Compared to the LPS/D-GalN group, the TAK-242 pretreatment group showed significantly prolonged survival, reduced serum alanine aminotransferase and aspartate aminotransferase levels, relieved oxidative stress, and reduced inflammatory interleukin (IL)-6, IL-12, and tumor necrosis factor-α levels. In addition, TAK-242 increased the accumulation of myeloid-derived suppressor cells (MDSCs). Next, mice were treated with an anti-Gr-1 antibody to deplete MDSCs, and adoptive transfer experiments were performed. We found that TAK-242 protected against FH by regulating MDSCs. In the in vitro studies, TAK-242 regulated the accumulation of MDSCs and promoted the release of immunosuppressive inflammatory cytokines. In addition, TAK-242 inhibited protein expression of nuclear factor-κB and mitogen-activated protein kinases. In summary, TAK-242 had a hepatoprotective effect against LPS/D-GalN-induced explosive hepatitis in mice. Its protective effect may be involved in suppressing inflammation, reducing oxidative stress, and increasing the proportion of MDSCs.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Liver/drug effects , Massive Hepatic Necrosis/prevention & control , Myeloid-Derived Suppressor Cells/drug effects , Protective Agents/therapeutic use , Sulfonamides/therapeutic use , Animals , Anti-Inflammatory Agents/pharmacology , Cytokines/metabolism , Drug Administration Schedule , Galactosamine , In Vitro Techniques , Lipopolysaccharides , Liver/immunology , Liver/metabolism , Male , Massive Hepatic Necrosis/etiology , Massive Hepatic Necrosis/immunology , Massive Hepatic Necrosis/metabolism , Mice , Mice, Inbred C57BL , Myeloid-Derived Suppressor Cells/immunology , Myeloid-Derived Suppressor Cells/metabolism , Oxidative Stress/drug effects , Protective Agents/pharmacology , Random Allocation , Sulfonamides/pharmacology , Treatment Outcome
9.
Int Immunopharmacol ; 89(Pt A): 107055, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33045579

ABSTRACT

Extracellular adenosine triphosphate (eATP) mediates pro-inflammatory responses by recruiting and activating inflammatory cells. CD39 can hydrolyze eATP into adenosine monophosphate (AMP), while CD73 can convert AMP into the immunosuppressive nucleoside adenosine (ADO). CD39 is a rate-limiting enzyme in this cascade, which is regarded as an immunological switch shifting the ATP-mediated pro-inflammatory environment to the ADO- mediated anti-inflammatory status. The CD39 expression can be detected in a wide spectrum of immunocytes, which is under the influence of environmental and genetic factors. It is increasingly suggested that, CD39 participates in some pathophysiological processes, like inflammatory bowel disease (IBD), sepsis, multiple sclerosis (MS), allergic diseases, ischemia-reperfusion (I/R) injury, systemic lupus erythematosus (SLE), diabetes and cancer. Here, we focus on the current understanding of CD39 in immunity, and comprehensively illustrate the diverse CD39 functions within a variety of disorders.


Subject(s)
Apyrase/metabolism , Gene Expression Regulation/immunology , Immunity, Cellular/physiology , Apyrase/genetics , Biomarkers , Drug Delivery Systems , Humans , Immune Checkpoint Proteins
10.
Int J Mol Sci ; 21(17)2020 Sep 02.
Article in English | MEDLINE | ID: mdl-32887217

ABSTRACT

Invasive breast cancer is highly regulated by tumor-derived cytokines in tumor microenvironment. The development of drugs that specifically target cytokines are promising in breast cancer treatment. In this study, we reported that arctigenin, a bioactive compound from Arctium lappa L., could decrease tumor-promoting cytokines GM-CSF, MMP-3, MMP-9 and TSLP in breast cancer cells. Arctigenin not only inhibited the proliferation, but also the invasion and stemness of breast cancer cells via decreasing GM-CSF and TSLP. Mechanistically, arctigenin decreased the promoter activities of GM-CSF and TSLP via reducing the nuclear translocation of NF-κB p65 which is crucial for the transcription of GM-CSF and TSLP. Furthermore, arctigenin-induced depletion of GM-CSF and TSLP inhibited STAT3 phosphorylation and ß-catenin signaling resulting in decreased proliferation, invasion and stemness of breast cancer cells in vitro and in vivo. Our findings provide new insights into the mechanism by which tumor-promoting cytokines regulate breast cancer progression and suggest that arctigenin is a promising candidate for cytokine-targeted breast cancer therapy.


Subject(s)
Breast Neoplasms/drug therapy , Cytokines/metabolism , Furans/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Lignans/pharmacology , STAT3 Transcription Factor/metabolism , beta Catenin/metabolism , Animals , Apoptosis , Biomarkers, Tumor , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Movement , Cell Proliferation , Cytokines/genetics , Female , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , STAT3 Transcription Factor/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , beta Catenin/genetics
12.
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
13.
Front Immunol ; 10: 1824, 2019.
Article in English | MEDLINE | ID: mdl-31428103

ABSTRACT

Macrophages play a critical role in the pathogenesis of endotoxin shock by producing excessive amounts of pro-inflammatory cytokines. A pan-caspase inhibitor, zVAD, can be used to induce necroptosis under certain stimuli. The role of zVAD in both regulating the survival and activation of macrophages, and the pathogenesis of endotoxin shock remains not entirely clear. Here, we found that treatment of mice with zVAD could significantly reduce mortality and alleviate disease after lipopolysaccharide (LPS) challenge. Notably, in LPS-challenged mice, treatment with zVAD could also reduce the percentage of peritoneal macrophages by promoting necroptosis and inhibiting pro-inflammatory responses in macrophages. In vitro studies showed that pretreatment with zVAD promoted LPS-induced nitric oxide-mediated necroptosis of bone marrow-derived macrophages (BMDMs), leading to reduced pro-inflammatory cytokine secretion. Interestingly, zVAD treatment promoted the accumulation of myeloid-derived suppressor cells (MDSCs) in a mouse model of endotoxin shock, and this process inhibited LPS-induced pro-inflammatory responses in macrophages. Based on these findings, we conclude that treatment with zVAD alleviates LPS-induced endotoxic shock by inducing macrophage necroptosis and promoting MDSC-mediated inhibition of macrophage activation. Thus, this study provides insights into the effects of zVAD treatment in inflammatory diseases, especially endotoxic shock.


Subject(s)
Amino Acid Chloromethyl Ketones/pharmacology , Caspase Inhibitors/pharmacology , Macrophage Activation/drug effects , Macrophages, Peritoneal/drug effects , Myeloid-Derived Suppressor Cells/drug effects , Necroptosis/drug effects , Shock, Septic/drug therapy , Animals , Apoptosis/drug effects , Caspases/metabolism , Female , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL , Myeloid-Derived Suppressor Cells/metabolism , Shock, Septic/metabolism
14.
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.

15.
J Cell Mol Med ; 23(7): 4738-4745, 2019 07.
Article in English | MEDLINE | ID: mdl-31062436

ABSTRACT

Long non-coding RNA MIR503 host gene (MIR503HG) is located on chromosome Xq26.3, and has been found to be deregulated in many types of human malignancy and function as tumour suppressor or promoter based on cancer types. The role of MIR503HG in breast cancer was still unknown. In our study, we found MIR503HG expression was significantly decreased in triple-negative breast cancer tissues and cell lines. Furthermore, we observed low MIR503HG expression was correlated with late clinical stage, lymph node metastasis and distant metastasis. In the survival analysis, we observed that triple-negative breast cancer patients with low MIR503HG expression had a statistically significant worse prognosis compared with those with high MIR503HG expression, and low MIR503HG expression was a poor independent prognostic factor for overall survival in triple-negative breast cancer patients. The study in vitro suggested MIR503HG inhibits cell migration and invasion via miR-103/OLFM4 axis in triple negative breast cancer. In conclusion, MIR503HG functions as a tumour suppressive long non-coding RNA in triple negative breast cancer.


Subject(s)
Cell Movement/genetics , Granulocyte Colony-Stimulating Factor/metabolism , MicroRNAs/metabolism , RNA, Long Noncoding/metabolism , Signal Transduction , Triple Negative Breast Neoplasms/genetics , Base Sequence , Cell Line, Tumor , Down-Regulation/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , MicroRNAs/genetics , Middle Aged , Multivariate Analysis , Neoplasm Invasiveness , Prognosis , Proportional Hazards Models , RNA, Long Noncoding/genetics
16.
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
17.
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
18.
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
19.
Am J Transl Res ; 10(5): 1552-1561, 2018.
Article in English | MEDLINE | ID: mdl-29887968

ABSTRACT

Cancer stem cells (CSCs) play important roles in tumor initiation, metastasis, and progression. They are also mainly responsible for high treatment failure rates. Identification and characterization of CSCs are crucial for facilitating the detection, prevention, or therapy of cancer. Great efforts have been paid to develop an effective method and the ideal method for CSCs research is still in the going. In our study, we created an ultra-low concentration of serum and non-adhesive (ULCSN) culture system to enrich CSCs from murine lewis lung cancer cell line LL/2 with cell spheres structure and characterize the LL/2 CSCs properties. Their characteristics were investigated through colony formation, spheres formation, chemoresistance, flow cytometry for putative stem cell markers, such as CD133, CD34 and CD45, immunofluorescence staining and tumor initiation capacity in vivo. Tumor spheres were formed within 7-10 days under the condition of ULCSN culture system. Compared with adherent parental LL/2 cells, the colony capacity, chemo-resistance, and expression of stem cell markers increased significantly in addition to tumor-initiating capability in the tumor sphere cells. Using the ULCSN culture system, an available isolation method of lewis lung CSCs was established, which is simple, effective, and inexpensive compared with the cytokines attachment serum free culture method. The stem cell properties of the tumor sphere LL/2 cells reflected the CSCs phenotypes. We developed a useful CSCs model for basic and pre-clinical studies for lung cancer and other types of cancer.

20.
Gerontology ; 64(5): 457-465, 2018.
Article in English | MEDLINE | ID: mdl-29804119

ABSTRACT

BACKGROUND: Tetrahydroxystilbene glucoside (TSG) is a main bioactive component of Polygonum multiflorum, a traditional Chinese medicine known for certain anti-aging effects. Since TSG has been found to extend lifespan in the nematode Caenorhabditis elegans, we hypothesized that TSG might produce anti-aging benefits in mammals. OBJECTIVE: The aim was to evaluate the anti-aging potential of TSG and to explore its relative molecular mechanism. METHODS: Mice were maintained on standard diet, high-calorie diet (HC), or high-calorie plus TSG diet. Survival rates and body weight changes were recorded weekly. Rotarod analysis was performed to assess the physical fitness of mice. Bone mineral density was assessed using micro-computed tomography. Hematoxylin and eosin staining was used for the histological examination of heart, liver, and kidney pathology. The mRNA and protein expression of target genes were analyzed by quantitative real-time polymerase chain reaction and western blotting, respectively. Mitotracker deep red staining and high-content analysis were used to quantify cellular mitochondrial mass and function. RESULTS: In this study, we found that TSG improved the physiology of aged mice consuming excess calories and delayed senile symptoms. The anti-aging benefits of TSG were mediated at least in part by the AMP-activated protein kinase (AMPK)/sirtuin 1 (SIRT1)/peroxisome proliferator-activated receptor-γ coactivator 1α (PGC-1α) signaling cascade, leading to significant improvement in motor function, bone mineral density, HC-induced organ pathology, and mitochondrial function. CONCLUSION: Our findings show that TSG could be a potential drug candidate for the treatment of aging- and high-calorie intake-associated disorders.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Aging/drug effects , Aging/metabolism , Glucosides/pharmacology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Sirtuin 1/metabolism , Stilbenes/pharmacology , Aging/pathology , Animals , Bone Density/drug effects , Cell Line , Drugs, Chinese Herbal/pharmacology , Energy Intake , Female , HeLa Cells , Humans , Mice , Mice, Inbred C57BL , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...