Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 78
Filter
Add more filters











Publication year range
1.
Mol Hum Reprod ; 27(11)2021 11 02.
Article in English | MEDLINE | ID: mdl-34643696

ABSTRACT

Evidence accumulated in recent years has revealed that neutrophils are involved in the initial establishment of endometriosis, which is well-known as a chronic inflammatory disease. So far, why and how neutrophils promote the formation of early endometriosis are still unclear. In this study, using a mouse model of endometriosis, we demonstrated that endometriosis mice (EMs mice) had a significantly increased number of neutrophils in peritoneal fluids and lesions, and increased levels of granulocyte colony-stimulating factor (G-CSF) and IL-6 in serum and peritoneal fluids compared to the control group. In the neutrophils and uterine fragments co-injection experiment, neutrophils regulated by G-CSF and IL-6 had a similar effect to neutrophils from EMs mice, increasing the number, area, weight and microvessel density (MVD) of endometriotic lesions. Blocking the effect of G-CSF and IL-6 in EMs mice resulted in a decrease in the number, area and weight of endometriotic lesions. Following the depletion of neutrophils in vivo using a anti-Ly6G antibody, the MVD in the lesions of mice treated with neutrophils from EMs mice and neutrophils from pG/pI6 mice were significantly reduced. Neutrophils from EMs mice and neutrophils from pG/pI6 mice altered the expression levels of Mmp9, Bv8 and Trail genes compared to the neutrophils from PBS-treated mice. IL-6 together with G-CSF induced a higher expression of phospho-STAT3 and STAT3 in neutrophils. These findings suggest that neutrophils modulated by G-CSF and IL-6 through the STAT3 pathway alter the expression levels of the angiogenesis-related genes Mmp9, Bv8 and Trail, and may promote the establishment of early endometriosis.


Subject(s)
Endometriosis/metabolism , Endometrium/blood supply , Endometrium/metabolism , Granulocyte Colony-Stimulating Factor/metabolism , Interleukin-6/metabolism , Neovascularization, Pathologic , Neutrophils/metabolism , Animals , Disease Models, Animal , Endometriosis/immunology , Endometriosis/pathology , Endometrium/immunology , Female , Gastrointestinal Hormones/genetics , Gastrointestinal Hormones/metabolism , Granulocyte Colony-Stimulating Factor/genetics , Interleukin-6/genetics , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Mice, Inbred BALB C , Microvascular Density , Neuropeptides/genetics , Neuropeptides/metabolism , Neutrophils/immunology , Phosphorylation , STAT3 Transcription Factor/metabolism , Signal Transduction , TNF-Related Apoptosis-Inducing Ligand/genetics , TNF-Related Apoptosis-Inducing Ligand/metabolism
2.
Mol Immunol ; 123: 106-115, 2020 07.
Article in English | MEDLINE | ID: mdl-32485469

ABSTRACT

Hepatocytes are the targets in autoimmune hepatitis (AIH) that results in T cell-dependent liver injury. However, hepatocytes may also affect the hepatic T cells in AIH, but the underlying mechanisms are not fully understood. Here we report that hepatocytes could secrete galectin-9 (Gal-9) to suppress the intrahepatic production of Th1 cytokine IFN-γ and restrict AIH development, but hepatocyte damage resulted in opposite effects due to release of TLR2/4 ligands that promoted the intrahepatic production of IL-1ß, IL-6, and IL-12. Through Tim-3, Gal-9 could efficiently suppress the intrahepatic T cell activation despite presence of TLR2/4 ligands, thus attenuating Th1 response in AIH. Intriguingly, intrahepatic IL-6/IL-12 suppressed the effect of TGF-ß on Treg cells. Therefore, in AIH, Gal-9 promoted Foxp3 expression and function of hepatic Treg cells through TL1A signaling, although Treg function was still impaired, compared with that in naive state. Due to its promoting effect on Treg function, together with its effect on T effector cells in a Tim-3-independent way, Gal-9 could attenuate intrahepatic IFN-γ production by hindering the increase of hepatic CD4+CD43+ T cells resulting from extrahepatic T cell activation. TLR2/4 ligands attenuated the effects of Gal-9 on Treg cells and CD4+CD43+ T cells by increasing intrahepatic IL-6 and IL-12. Blocking TLR2/4 ligands could efficiently suppress intrahepatic IFN-γ production, liver injury, and hepatic fibrosis. These findings suggest that hepatocytes paradoxically affect Th1 response in AIH due to Gal-9 expression and TLR2/4 ligands release, and that targeting TLR2/4 signaling may provide an important approach in the therapeutic strategy for AIH.


Subject(s)
Galectins/metabolism , Hepatitis, Autoimmune/metabolism , Hepatocytes/physiology , Interferon-gamma/metabolism , Liver/metabolism , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/metabolism , Animals , Cells, Cultured , Hepatitis, Autoimmune/immunology , Hepatitis, Autoimmune/pathology , Ligands , Liver/pathology , Mice , Mice, Inbred C57BL , Signal Transduction , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/pathology , Th1 Cells/immunology , Th1 Cells/metabolism
3.
Mol Hum Reprod ; 26(4): 240-255, 2020 04 24.
Article in English | MEDLINE | ID: mdl-32119739

ABSTRACT

Endometriosis is a gynecological disease with abnormal expression of interleukin (IL)-37 which can suppress inflammation and the immune system. Here we investigated the role of the IL-37b splice variant in endometriosis in vivo and in vitro. In a murine model of endometriosis, in vivo administration of IL-37b significantly inhibited the development of lesions judged by the number (P = 0.0213), size (P = 0.0130) and weight (P = 0.0152) of lesions. IL-37b had no effect on the early stage of lesion formation, however administration in the growth stage of lesions decreased the number (P = 0.0158), size (P = 0.0158) and weight (P = 0.0258) of lesions compared with PBS control, an effect that was not reversed by macrophage depletion. Expressions of inflammatory factors, matrix metalloproteinases and vascular endothelial growth factor-A mRNA/protein were significantly inhibited in ectopic lesions following IL-37b administration, and in uterine segments treated in vitro. In vitro treatment of uterine segments with IL-37b inhibited phosphorylation of Akt and Erk1/2 in uterine segments. Isolated mouse endometrial stromal treated with IL-37b and transfected with pIL-37b plasmid got suppressed cell proliferation, invasion, angiogenesis and the expression of inflammatory factors. In addition, transfection with pIL-37b significantly decreased the phosphorylation of Akt and Erk1/2. IL-37b also inhibited proliferation and the expression of inflammatory and angiogenesis factors in epithelial cell line RL95-2. These findings suggest that IL-37b may inhibit the growth of lesions by regulating proliferation, invasion, angiogenesis and inflammation through Akt and Erk1/2 signaling pathway.


Subject(s)
Endometriosis/prevention & control , Interleukin-1/pharmacology , Animals , Cell Proliferation/drug effects , Disease Models, Animal , Endometriosis/drug therapy , Female , Inflammation/prevention & control , MAP Kinase Signaling System/drug effects , Macrophages, Peritoneal/drug effects , Mice , Mice, Inbred BALB C , Neovascularization, Pathologic/prevention & control , Proto-Oncogene Proteins c-akt/antagonists & inhibitors
4.
J Transl Med ; 18(1): 3, 2020 01 06.
Article in English | MEDLINE | ID: mdl-31906950

ABSTRACT

BACKGROUND: The prevalence of autoimmune hepatitis (AIH) is increasing, and its early clinical diagnosis is difficult. The pathogenesis of AIH remains unclear, and AIH-related studies are largely limited because of lack of suitable mouse models. METHODS: To obtain a good tool for research on AIH, we first established an improved immune-mediated mouse model that can mimic the pathological process of AIH as in the human body, through repeated injections of human cytochrome P450 2D6 (CYP2D6) plasmid. Next, a proteomic analysis based on isobaric tag (IBT) technology was performed to detect the differentially expressed proteins (DEPs), and related biological functions and pathways in the plasma of AIH and normal mice. Finally, we performed enzyme-linked immunosorbent assay (ELISA) to further confirm the most abundant DEP in the plasma of patients with AIH. RESULTS: Autoantibodies and the characteristic pathology of AIH were observed in our mouse model. Inflammatory infiltration also increased in the livers of AIH mice over time and plateaued by day 42 post the first injection. Chronic hepatitis was most severe on day 35 with the development of fibrosis as well, and the plasma of AIH mice were collected for proteomic analysis. A total of 176 DEPs were found in this experiment, of which 148 DEPs were up-regulated and 28 DEPs were down-regulated. Thirty significant Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways (P < 0.05) were detected. Arginine biosynthesis was found to be the most significant pathway involved in the AIH process. During the Gene Ontology (GO) analysis, most DEPs were found to be involved in the binding, cellular, and metabolic processes. Using ELISA, the most overexpressed DEP, serum amyloid A 1 (SAA1), was confirmed to be increased specifically in the plasma of patients with AIH compared to other chronic hepatitis. Different plasma levels of SAA1 were also found related to different grades of inflammation and stages of fibrosis in the liver of patients with AIH. CONCLUSIONS: Our study is the first to describe the proteomics analysis of a true sense of AIH mouse model, which is beneficial for a better understanding of AIH pathogenesis and identifying potential biomarkers for its clinical diagnosis.


Subject(s)
Hepatitis, Autoimmune , Animals , Autoantibodies , Autoantigens , Disease Models, Animal , Humans , Mice , Proteomics
6.
Immunol Lett ; 218: 44-50, 2020 02.
Article in English | MEDLINE | ID: mdl-31794800

ABSTRACT

Autoimmune hepatitis (AIH) is a chronic liver disease mediated by immunity, and could lead to liver fibrosis and hepatocellular carcinoma. However, the mechanisms for breaking hepatic tolerance and driving AIH still remain elusive. We herein reported that the non-specific liver inflammation triggered by carbon tetrachloride (CCl4) recruited high numbers of CD4+T, CD8+T and B cells, and elevated the expression of proinflammaitory cytokines in Balb/c mice, further breaking liver tolerance and inducing autoimmune response, AIH inflammation and liver fibrosis in the presence of CYP2D6 antigen mimicry. In contrast, adenovirus infection could not break liver tolerance and induce AIH in Balb/c mice even in the presence of CYP2D6 antigen mimicry. These results suggested that genetic predisposition could determine liver tolerance in Balb/c mice. The chemical induced inflammation in the liver breaks tolerance and might be considered important for the initiation and development of AIH in Balb/c mice.


Subject(s)
Autoimmunity , Disease Susceptibility , Hepatitis, Autoimmune/etiology , Immune Tolerance , Adenoviridae/immunology , Animals , Autoantigens/immunology , Biomarkers , Biopsy , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/pathology , Carbon Tetrachloride/adverse effects , Disease Models, Animal , Disease Susceptibility/immunology , Female , Hepatitis, Autoimmune/metabolism , Hepatitis, Autoimmune/pathology , Immunohistochemistry , Mice , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/pathology
9.
Int Immunopharmacol ; 69: 184-193, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30735937

ABSTRACT

T cell-dependent liver injury is an important reason for the massive hepatic damage and cirrhosis. So far it is unclear whether the development of the disease could be efficiently suppressed by anti-inflammatory cytokine that modulates innate immune cells. Here we report that anti-inflammatory cytokine IL-37 could efficiently suppress the sustained hepatic expression of IFN-γ and TNF-α, two critical cytokines for inducing hepatocyte apoptosis and liver fibrosis in T cell-dependent liver injury. IL-37 could directly suppress IFN-γ/TLR4 ligand-induced M1 activation of macrophages, thus reducing the expression of pro-inflammatory cytokines TNF-α, IL-1ß, and IL-12. Moreover, IL-37 attenuated Th1 response in vivo and increased the expression of Th2 cytokines IL-4 and IL-13, which in turn promoted M2 activation of macrophages in the liver. The increase of M2 activation not only further reduced TNF-α, IL-1ß and IL-12 expression, but also increased IL-10 and IL-1Ra expression in macrophages, thus more efficiently suppressing the hepatic IFN-γ expression. By suppressing IFN-γ/TNF-α expression, IL-37 suppressed the up-regulation and activation of MLKL that drives hepatocellular necrosis in T cell-dependent liver damage. Accordingly, IL-37 efficiently reduced liver injury and hepatic inflammation after the repeated ConA challenge and the induction of autoimmune hepatitis, and also suppressed hepatic fibrosis resulting from the sustained liver damage. This study showed that the direct and indirect effect of IL-37 on macrophages could reduce the hepatic TNF-α expression, and also modulate IL-1ß/IL-12 and IL-10/IL-1Ra expression to suppress the hepatic IFN-γ expression, thus suppressing the development of T cell-dependent liver injury such as autoimmune hepatitis.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Hepatitis, Autoimmune/drug therapy , Hepatocytes/physiology , Interleukin-1/therapeutic use , Macrophages/physiology , T-Lymphocytes/immunology , Animals , Cells, Cultured , Concanavalin A , Cytokines/metabolism , Disease Models, Animal , Humans , Immunosuppression Therapy , Interferon-gamma/metabolism , Mice , Receptors, Interleukin-10/metabolism , Tumor Necrosis Factor-alpha/metabolism
10.
J Cell Commun Signal ; 13(3): 369-380, 2019 Sep.
Article in English | MEDLINE | ID: mdl-30739244

ABSTRACT

TGF-ß1 is a main inducer of epithelial to mesenchymal transition (EMT). However, many breast cancer cells are not sensitive to the EMT induction by TGF-ß1 alone. So far, the mechanisms underlying the induction of TGF-ß1-insensitive breast cancer cells remains unclear. Here we report that TNF-α can induce EMT and invasiveness of breast cancer cells which are insensitive to TGF-ß1. Intriguingly, TGF-ß1 could cooperate with TNF-α to promote the EMT and invasiveness of breast cancer cells. The prolonged co-stimulation with TGF-ß1 and TNF-α could enhance the sustained activation of Smad2/3, p38 MAPK, ERK, JNK and NF-κB pathways by enhancing the activation of TAK1, which was mediated by the gradually up-regulated TßRs. Except for JNK, all of these pathways were required for the effects of TGF-ß1 and TNF-α. Importantly, the activation of p38 MAPK and ERK pathways resulted in a positive feed-back effect on TAK1 activation by up-regulating the expression of TßRs, favoring the activation of multiple signaling pathways. Moreover, SLUG was up-regulated and required for the TGF-ß1/TNF-α-induced EMT and invasiveness. In addition, SLUG could also enhance the activation of signaling pathways by promoting TßRII expression. These findings suggest that the up-regulation of TßRs contributes to the sustained activation of TAK1 induced by TGF-ß1/TNF-α and the following activation of multiple signaling pathways, resulting in EMT and invasiveness of breast cancer cells.

11.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 34(8): 690-694, 2018 Aug.
Article in Chinese | MEDLINE | ID: mdl-30384866

ABSTRACT

Objective To investigate the effect of human cytochrome P450 family 2 subfamily D member 6 (CYP2D6) on the establishment of animal model of autoimmune hepatitis (AIH) in C57BL/6 and BALB/c mice. Methods Adenovirus (Ad) and plasmid pCYP2D6 were injected into the tail vein of mice. Serum anti-CYP2D6 antibody and hepatic protein (HP) autoantibody were detected by ELISA. HE staining was used to detect the degree of liver AIH inflammation, and the degree of liver fibrosis was detected by sirius red staining. The level of alanine aminotransferase (ALT) was determined by microplate method. Results Ad and self-mimicking antigen (human CYP2D6) induced the secretion of HP autoantibody, AIH, hepatic injury and liver fibrosis in C57BL/6 mice, whereas only anti-CYP2D6 antibody was generated in BALB/c mice; no HP autoantibody, AIH or liver fibrosis was induced. Conclusion In the presence of AD, the continuous expression of self-mimicking antigen (human CYP2D6) could induce AIH in C57BL/6 mice, but not in BALB/c mice. Thus, the genetic background in the different strains of mice could influence the occurrence and development of AIH.


Subject(s)
Hepatitis, Autoimmune , Animals , Autoantibodies , Autoantigens , Cytochrome P-450 CYP2D6 , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL
12.
J Alzheimers Dis ; 65(4): 1385-1400, 2018.
Article in English | MEDLINE | ID: mdl-30175975

ABSTRACT

BACKGROUND: Axonopathy is closely linked to the development of diabetic encephalopathy induced by type II diabetes (T2D). Berberine has been shown to cross the blood-brain barrier and holds promising effect for neuronal damage in diabetes. OBJECTIVE: The present study investigated the protective effect and the underlying mechanism of berberine on neuronal axonopathy in both in vitro and in vivo models. METHODS: High glucose/high fat diet and streptozotocin injection-induced T2D rat model was used. Berberine was administered p.o. to T2D rat model for 10 weeks. Morris water maze test, in vivo neuronal tracing, immunohistochemistry, and western blot analysis were performed to evaluate the protective effects of berberine in T2D-induced diabetic encephalopathy rats. Primary cultured neurons were used to further explore the underlying mechanisms in vitro. RESULTS: Berberine dramatically reduced blood glucose and serum insulin levels and alleviated insulin resistance. Berberine significantly attenuated memory impairment, axonopathy, and tau hyperphosphorylation, and also restored PI3K/Akt/GSK3ß signaling pathway in T2D rats. In vitro, berberine induced an increase in the phosphorylation of PI3K/Akt as well as GSK3ß in high glucose-treated primary neurons. Furthermore, berberine-induced PI3K/Akt activation also resulted in the dephosphorylation of tau protein, which could improve axonal transport impairment in high glucose-treated primary neurons. Pretreated neurons with LY294002, an inhibitor of PI3K, partially blocked berberine-inhibited tau phosphorylation and berberine-activated PI3K/Akt signaling pathway. CONCLUSIONS: Berberine exerts the protective effect against cognitive deficits by improving tau hyperphosphorylation and the axonal damage through restoring PI3K/Akt/GSK3ß signaling pathway.


Subject(s)
Axons/pathology , Berberine/therapeutic use , Diabetic Nephropathies/drug therapy , Signal Transduction/physiology , tau Proteins/metabolism , Animals , Axons/drug effects , Berberine/pharmacology , Cells, Cultured , Chromones/pharmacology , Diabetes Mellitus, Type 2/chemically induced , Diabetes Mellitus, Type 2/complications , Diabetic Nephropathies/blood , Disease Models, Animal , Embryo, Mammalian , Enzyme Inhibitors/pharmacology , Female , Glucose/pharmacology , Male , Morpholines/pharmacology , Pregnancy , Rats , Rats, Wistar , Signal Transduction/drug effects , Streptozocin
13.
Nat Commun ; 9(1): 1739, 2018 04 30.
Article in English | MEDLINE | ID: mdl-29712898

ABSTRACT

Chemoresistance is a major unmet clinical obstacle in ovarian cancer treatment. Epigenetics plays a pivotal role in regulating the malignant phenotype, and has the potential in developing therapeutically valuable targets that improve the dismal outcome of this disease. Here we show that a series of transcription factors, including C/EBPß, GCM1, and GATA1, could act as potential modulators of histone methylation in tumor cells. Of note, C/EBPß, an independent prognostic factor for patients with ovarian cancer, mediates an important mechanism through which epigenetic enzyme modifies groups of functionally related genes in a context-dependent manner. By recruiting the methyltransferase DOT1L, C/EBPß can maintain an open chromatin state by H3K79 methylation of multiple drug-resistance genes, thereby augmenting the chemoresistance of tumor cells. Therefore, we propose a new path against cancer epigenetics in which identifying and targeting the key regulators of epigenetics such as C/EBPß may provide more precise therapeutic options in ovarian cancer.


Subject(s)
Biomarkers, Tumor/genetics , CCAAT-Enhancer-Binding Protein-beta/genetics , Chromatin/metabolism , Drug Resistance, Neoplasm/genetics , Epigenesis, Genetic , Histones/genetics , Ovarian Neoplasms/genetics , Antineoplastic Agents/pharmacology , Biomarkers, Tumor/metabolism , CCAAT-Enhancer-Binding Protein-beta/metabolism , Cell Line, Tumor , Chromatin/chemistry , Cisplatin/pharmacology , DNA-Binding Proteins , Databases, Factual , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , GATA1 Transcription Factor/genetics , GATA1 Transcription Factor/metabolism , GATA4 Transcription Factor/genetics , GATA4 Transcription Factor/metabolism , Histone-Lysine N-Methyltransferase , Histones/metabolism , Humans , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/metabolism , Methylation , Methyltransferases/genetics , Methyltransferases/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/mortality , Ovarian Neoplasms/pathology , Primary Cell Culture , Prognosis , Survival Analysis , Transcription Factors/genetics , Transcription Factors/metabolism
14.
Mol Immunol ; 99: 171-181, 2018 07.
Article in English | MEDLINE | ID: mdl-29793131

ABSTRACT

Autoimmune hepatitis (AIH), a serious autoimmune liver disease, can be a lifelong illness, leading to fibrosis, cirrhosis, and hepatocellular carcinoma (HCC). So far the mechanisms for disease initiation are largely unknown. Here we report that the amplified non-AIH liver inflammation could promote the initiation of AIH due to the sustained increase of IL-6, IL-12, IL-4, and IL-25 in the liver. The liver injury resulting from virus (adenovirus) or chemicals (CCl4) could induce an amplified (stronger/long-lasting) hepatic inflammation by releasing the ligands for TLR2/TLR4. The amplified inflammation resulted in the increase of multiple cytokines and chemokines in the liver. Among them, the sustained increase of IL-6/IL-12 resulted in the activation of STAT3 and STAT4 in hepatic CD4+CD25+ Treg cells, thus suppressing Foxp3 gene expression to reduce the suppressive function of Treg cells in the liver, but not those in the spleen. The increase of IL-12 and the impairment of Treg function promoted Th1 response in presence of self-mimicking antigen (human CYP2D6). Intriguingly, the amplified inflammation resulted in the increase of IL-4 and IL-25 in the liver. The moderate increase of IL-4 was sufficient for cooperating with IL-25 to initiate Th2 response, but inefficient in suppressing Th1 response, favoring the initiation of autoimmune response. Consequently, either adenovirus/CYP2D6 or CCl4/CYP2D6 could induce the autoimmune response and AIH in the mice, leading to hepatic fibrosis. The findings in this study suggest that the amplified non-AIH inflammation in the liver could be a driving force for the initiation of autoimmune response and AIH.


Subject(s)
Autoimmunity/immunology , Hepatitis, Autoimmune/immunology , Inflammation/immunology , Liver/immunology , Toll-Like Receptor 2/immunology , Toll-Like Receptor 4/immunology , Animals , Female , Interleukins/immunology , Ligands , Mice , Mice, Inbred C57BL , T-Lymphocytes, Regulatory/immunology
15.
Oncotarget ; 8(20): 33501-33514, 2017 May 16.
Article in English | MEDLINE | ID: mdl-28432279

ABSTRACT

IL-35 is an immunosuppressive cytokine and exerts regulatory effects on T cells, B cells, macrophages and dendritic cells. Neutrophils are important innate immune cells that play key roles in tumor development. The effect of IL-35 on neutrophils remains unknown. Here, we report that IL-35 can induce N2 neutrophil polarization (protumor phenotype) by increasing G-CSF and IL-6 production, and promote neutrophil infiltration into tumor microenvironment. The sustained expression of IL-35 could promote chronic inflammation to augment the proangiogenic and immunosuppressive function of neutrophils. IL-35 stimulated macrophages to secrete proinflammatory cytokines IL-1ß and IL-6. IL-1ß stimulated γδ T cells to produce IL-17, which in turn increased the production of G-CSF. By increasing the expression of G-CSF and IL-6, IL-35 could up-regulate the expression of MMP-9 and Bv8, and down-regulate TRAIL expression in neutrophils, thus augmenting the proangiogenic function of neutrophils. Moreover, G-CSF/IL-6 induced the enhanced activation of STAT3 and ERK pathways in neutrophils, thus increasing the expression of iNOS to suppress T cell activation. Our findings suggest that IL-35 can promote tumor progression by functioning as an up-stream cytokine to promote cancer-associated inflammation and control neutrophil polarization. Targeting IL-35 might be an important approach for designing new strategy of tumor therapy.


Subject(s)
Interleukins/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Neutrophil Infiltration , Neutrophils/metabolism , Phenotype , Animals , Biomarkers , Cytokines/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Immunomodulation , Interleukins/pharmacology , Macrophages/immunology , Macrophages/metabolism , Macrophages/pathology , Mice , Neoplasms/immunology , Neovascularization, Pathologic/immunology , Neovascularization, Pathologic/metabolism , Neutrophil Infiltration/immunology , Neutrophils/drug effects , Neutrophils/immunology , Neutrophils/pathology , Nitric Oxide Synthase Type II/metabolism , STAT3 Transcription Factor/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
16.
Oncotarget ; 8(17): 28418-28430, 2017 Apr 25.
Article in English | MEDLINE | ID: mdl-28415700

ABSTRACT

Circulating tumor cells (CTCs) have been studied well in the prognosis for malignant diseases as liquid biopsy, but their contribution to tumor metastasis is not clearly defined. Here we report that CTCs could promote the metastatic colonization of disseminated carcinoma cells by inducing systemic inflammation and neutrophil recruitment to pre-metastatic organs. Depletion of neutrophils in vivo could effectively abrogate the promoting effect of CTCs on tumor cell metastasis. In the presence of CTCs, the pro-tumor function of neutrophils was augmented, whereas the antitumor function of neutrophils was suppressed. Mechanically, CTC-derived ligands for TLR2 and TLR4 (TLR2/4) induced the systemic inflammation, thus increasing the production of proinflammatory cytokines such as G-CSF and IL-6 that could induce the conversion of neutrophil function from tumor-suppressing to tumor-promoting. Moreover, CTCs induced the production of endogenous TLR2/4 ligands such as S100A8, S100A9, and SAA3, which may amplify the stimulating effect that induces the expression of proinflammatory cytokines. The promoting effect of CTCs on tumor cell metastasis could be abrogated by suppressing inflammatory response with IL-37, an anti-inflammatory cytokine, or blocking CTC-derived ligands for TLR2/4. Identification of the metastatic axis of CTCs/systemic inflammation/neutrophils may provide potential targets for preventing tumor cell metastasis.


Subject(s)
Inflammation/pathology , Neoplasms/pathology , Neoplastic Cells, Circulating/pathology , Animals , Biomarkers , Cell Degranulation/genetics , Cell Degranulation/immunology , Cytokines/metabolism , Disease Models, Animal , Gene Expression , Inflammation/etiology , Inflammation/metabolism , Inflammation Mediators/metabolism , Ligands , Melanoma, Experimental , Mice , Neoplasm Metastasis , Neoplasms/complications , Neoplasms/genetics , Neoplasms/metabolism , Neoplastic Cells, Circulating/metabolism , Neutrophil Infiltration , Neutrophils/immunology , Neutrophils/metabolism , Neutrophils/pathology , TNF-Related Apoptosis-Inducing Ligand/genetics , TNF-Related Apoptosis-Inducing Ligand/metabolism , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/metabolism
17.
Clin Cancer Res ; 23(1): 214-224, 2017 Jan 01.
Article in English | MEDLINE | ID: mdl-27401250

ABSTRACT

PURPOSE: Lymphatic vessels are mainly regarded as passive conduits for the dissemination of cancer cells. In this study, we investigate whether and how the tumor-associated lymphatic vessels may play an active role in tumor metastasis. EXPERIMENTAL DESIGN: In situ laser capture microdissection of lymphatic vessels followed by cDNA microarray analysis was used to determine the expression profiling of lymphatic endothelial cells (LEC). Gene expression levels and activity of signaling pathways were measured by real-time RT-PCR, ELISA, or immunoblotting. Lymphangiogenesis was assessed by IHC. Lymph node metastasis was measured using fluorescence imaging. The effects of SEMA4C on lymphangiogenesis in vitro were evaluated using migration assay and tube-formation assay of LECs. RESULTS: Tumor-associated LECs are molecularly and functionally different from their normal counterparts. In addition to expressing high levels of membrane-bound SEMA4C, tumor-associated LECs also produced soluble SEMA4C (sSEMA4C). Increased serum sSEMA4C was detected in patients with breast cancer and cervical cancer. Patients with metastasis had much higher levels of serum sSEMA4C. sSEMA4C promoted lymphangiogenesis by activating PlexinB2-ERBB2 signaling in LECs, and promoted the proliferation and migration of tumor cells by activating PlexinB2-MET signaling, thus promoting lymphatic metastasis. Although the SEMA4C signaling pathways differ between LECs and tumor cells, RHOA activation was necessary for the effects of SEMA4C in both types of cells. CONCLUSIONS: Tumor-associated LECs produce sSEMA4C to promote lymphatic metastasis of tumors. Our results suggest that SEMA4C and RHOA might be potential therapeutic targets, and that higher serum sSEMA4C could be a marker for breast cancer and cervical cancer. Clin Cancer Res; 23(1); 214-24. ©2016 AACR.


Subject(s)
Endothelial Cells/metabolism , Gene Expression Regulation, Neoplastic , Neoplasms/genetics , Neoplasms/metabolism , Semaphorins/genetics , Semaphorins/metabolism , Animals , Cell Line, Tumor , Cell Movement , Cell Proliferation , Disease Models, Animal , Female , Gene Expression Profiling , Heterografts , Humans , Immunohistochemistry , Lymphangiogenesis , Lymphatic Metastasis , Lymphatic Vessels/metabolism , Lymphatic Vessels/pathology , Mice, Nude , Neoplasms/pathology , Receptor, ErbB-2/metabolism , Signal Transduction , rhoA GTP-Binding Protein/metabolism
18.
Oncotarget ; 7(28): 43324-43336, 2016 Jul 12.
Article in English | MEDLINE | ID: mdl-27270316

ABSTRACT

Infiltrating neutrophils are known to promote in the development of tumor. However, it is unclear whether and how neutrophils are involved in triggering the growth of dormant metastases. Here we show that 14,15-epoxyeicosatrienoic acid (14,15-EET) can trigger the growth of dormant micrometastases by inducing neutrophilic infiltration and converting neutrophil function. 14,15-EET triggered neutrophil infiltration in metastatic lesions by activating STAT3 and JNK pathways to induce the expression of human IL-8 and murine CXCL15 in corresponding tumor cells. The continuous expression of hIL-8/mCXCL15 was maintained by the sustained and enhanced activation of JNK pathway. 14,15-EET up-regulated miR-155 expression by activating STAT3 and JNK pathways. miR-155 in turn down-regulated the expression of SHIP1 and DET1, thus augmenting the activation of JNK and c-Jun. Moreover, the function of neutrophils was converted from tumor-suppressing to tumor-promoting by 14,15-EET in vivo. By inducing the production of G-CSF/IL-6 in vivo, 14,15-EET induced the enhancement of STAT3 activation in neutrophils to increase MMP-9 expression and decrease TRAIL expression. Neutrophil-derived MMP-9 was required for 14,15-EET to induce angiogenesis during the growth of dormant micrometastases. Depleting neutrophils or inhibiting hIL-8/mCXCL15 up-regulation resulted in the failure of 14,15-EET to promote the development of micrometastases. These findings reveal a mechanism through which the infiltration and tumor-promoting function of neutrophils could be induced to trigger the growth of dormant metastases, which might be a driving force for the tumor recurrence based on dormant metastases.


Subject(s)
8,11,14-Eicosatrienoic Acid/analogs & derivatives , Neoplasm Micrometastasis/pathology , Neoplasm Recurrence, Local/pathology , Neovascularization, Pathologic/pathology , Neutrophil Infiltration/drug effects , Neutrophils/pathology , 8,11,14-Eicosatrienoic Acid/pharmacology , Animals , Chemokines, CXC/genetics , Chemokines, CXC/metabolism , Down-Regulation , Granulocyte Colony-Stimulating Factor/metabolism , Hep G2 Cells , Humans , Interleukin-6/metabolism , Interleukin-8/genetics , Interleukin-8/metabolism , MCF-7 Cells , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred C57BL , Mice, Nude , MicroRNAs/metabolism , Neoplasm Invasiveness/pathology , Neutrophils/metabolism , RNA Interference , RNA, Small Interfering/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Up-Regulation , Xenograft Model Antitumor Assays
20.
Cancer Sci ; 106(5): 559-66, 2015 May.
Article in English | MEDLINE | ID: mdl-25683165

ABSTRACT

Sirtuin 6 (SIRT6) can function as a tumor suppressor by suppressing aerobic glycolysis and apoptosis resistance. However, the negative effect of SIRT6 on cellular senescence implies that it may also have the potential to promote tumor development. Here we report that the upregulation of SIRT6 expression was required for transforming growth factor (TGF)-ß1 and H2O2/HOCl reactive oxygen species (ROS) to promote the tumorigenicity of hepatocellular carcinoma (HCC) cells. Transforming growth factor-ß1/H2O2/HOCl could upregulate SIRT6 expression in HCC cells by inducing the sustained activation of ERK and Smad pathways. Sirtuin 6 in turn abrogated the inducing effect of TGF-ß1/H2O2/HOCl on cellular senescence of HCC cells, and was required for the ERK pathway to efficiently suppress the expression of p16 and p21. Sirtuin 6 altered the effect of Smad and p38 MAPK pathways on cellular senescence, and contributed to the inhibitory effect of the ERK pathway on cellular senescence. However, SIRT6 was inefficient in antagonizing the promoting effect of TGF-ß1/H2O2 HOCl on aerobic glycolysis and anoikis resistance. Intriguingly, if SIRT6 expression was inhibited, the promoting effect of TGF-ß1/H2O2/HOCl on aerobic glycolysis and anoikis resistance was not sufficient to enhance the tumorigenicity of HCC cells. Suppressing the upregulation of SIRT6 enabled TGF-ß1/H2O2/HOCl to induce cellular senescence, thereby abrogating the enhancement of HCC cell tumorigenicity by TGF-ß1/H2O2/HOCl. These results suggest that SIRT6 is required for TGF-ß1/H2O2/HOCl to enhance the tumorigenicity of HCC cells, and that targeting the ERK pathway to suppress the upregulation of SIRT6 might be a potential approach in comprehensive strategies for the therapy of HCC.


Subject(s)
Carcinoma, Hepatocellular/pathology , Cellular Senescence/drug effects , Liver Neoplasms/pathology , Sirtuins/metabolism , Transforming Growth Factor beta1/metabolism , Animals , Anoikis , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Glycolysis , Humans , Hydrogen Peroxide/metabolism , Hydrogen Peroxide/pharmacology , Hypochlorous Acid/metabolism , Hypochlorous Acid/pharmacology , Liver Neoplasms/metabolism , MAP Kinase Signaling System , Mice, Nude , Sirtuins/genetics , Smad Proteins/metabolism , Transforming Growth Factor beta1/pharmacology , Up-Regulation , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL